WO2020081568A1 - Programmable dna base editing by nme2cas9-deaminase fusion proteins - Google Patents

Programmable dna base editing by nme2cas9-deaminase fusion proteins Download PDF

Info

Publication number
WO2020081568A1
WO2020081568A1 PCT/US2019/056341 US2019056341W WO2020081568A1 WO 2020081568 A1 WO2020081568 A1 WO 2020081568A1 US 2019056341 W US2019056341 W US 2019056341W WO 2020081568 A1 WO2020081568 A1 WO 2020081568A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
nme2cas9
editing
deaminase
virus
Prior art date
Application number
PCT/US2019/056341
Other languages
French (fr)
Inventor
Erik J. SONTHEIMER
Xin Gao
Aamir MIR
Alireza EDRAKI
Scot A. Wolfe
Pengpeng LIU
Original Assignee
University Of Massachusetts
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to CA3116555A priority Critical patent/CA3116555A1/en
Priority to US17/285,440 priority patent/US20220290113A1/en
Priority to MX2021004301A priority patent/MX2021004301A/en
Priority to SG11202103722TA priority patent/SG11202103722TA/en
Priority to CN201980077403.7A priority patent/CN113166743A/en
Priority to EP19806344.8A priority patent/EP3867367A1/en
Priority to BR112021007123-7A priority patent/BR112021007123A2/en
Priority to AU2019362874A priority patent/AU2019362874A1/en
Application filed by University Of Massachusetts filed Critical University Of Massachusetts
Priority to EA202191033A priority patent/EA202191033A1/en
Priority to KR1020217014669A priority patent/KR20210077732A/en
Priority to JP2021545287A priority patent/JP2022508716A/en
Publication of WO2020081568A1 publication Critical patent/WO2020081568A1/en
Priority to IL282267A priority patent/IL282267A/en
Priority to CONC2021/0006336A priority patent/CO2021006336A2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • C12N9/22Ribonucleases RNAses, DNAses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • C12N15/625DNA sequences coding for fusion proteins containing a sequence coding for a signal sequence
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/87Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
    • C12N15/90Stable introduction of foreign DNA into chromosome
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/78Hydrolases (3) acting on carbon to nitrogen bonds other than peptide bonds (3.5)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/04Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in cyclic amidines (3.5.4)
    • C12Y305/04004Adenosine deaminase (3.5.4.4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y305/00Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5)
    • C12Y305/04Hydrolases acting on carbon-nitrogen bonds, other than peptide bonds (3.5) in cyclic amidines (3.5.4)
    • C12Y305/04005Cytidine deaminase (3.5.4.5)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/09Fusion polypeptide containing a localisation/targetting motif containing a nuclear localisation signal
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/20Type of nucleic acid involving clustered regularly interspaced short palindromic repeats [CRISPRs]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14141Use of virus, viral particle or viral elements as a vector
    • C12N2750/14142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2750/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssDNA viruses
    • C12N2750/00011Details
    • C12N2750/14011Parvoviridae
    • C12N2750/14111Dependovirus, e.g. adenoassociated viruses
    • C12N2750/14171Demonstrated in vivo effect
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/80Vectors containing sites for inducing double-stranded breaks, e.g. meganuclease restriction sites

Definitions

  • the present invention is related to the field of gene editing.
  • the gene editing is directed toward single nucleotide base editing.
  • single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair.
  • the high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein.
  • the compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein to have a gene editing window that can edit sites that are not targetable by other conventional SpyCas9 base editor platforms.
  • CRISPR Clustered regularly interspaced short palindromic repeats
  • Cas CRISPR associated proteins
  • SpyCas9 base editing platforms cannot be used to target all single-base mutations due to their limited editing windows.
  • the editing window is constrained in part by the requirement for an NGG PAM and by the requirement that the edited base(s) be a very precise distance from the PAM.
  • SpyCas9 is also intrinsically associated with high off-targeting effects in genome editing.
  • the present invention is related to the field of gene editing.
  • the gene editing is directed toward single nucleotide base editing.
  • single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair.
  • the high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein.
  • the compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein to have a gene editing window that is superior to other conventional SpyCas9 base editor platforms.
  • the present invention contemplates a mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N 4 CC nucleotide sequence.
  • said protein is Nme2Cas9.
  • said protein further comprises a nuclear localization signal protein.
  • said nucleotide deaminase is a cytidine deaminase.
  • said nucleotide deaminase is an adenosine deaminase.
  • the protein further comprises a uracil glycosylase inhibitor.
  • the said nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS.
  • said binding region is a protospacer accessory motif interacting domain.
  • said protospacer accessory motif interacting domain comprises said mutation.
  • said mutation is a D16A mutation.
  • said mutated NmeCas9 protein further comprises CBE4.
  • said mutated NmeCas9 protein further comprises a linker.
  • said linker is a 73aa linker.
  • said linker is a 3xHA-tag.
  • the present invention contemplates a construct, wherein said construct is an optimized nNme2Cas9-ABEmax.
  • the present invention contemplates a construct, wherein said construct is a nNme2Cas9-CBE4.
  • the present invention contemplates a construct, wherein said construct is a YE1-BE3-nNme2Cas9 (D16A)-UGI.
  • the present invention contemplates an adeno-associated virus comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N 4 CC nucleotide sequence.
  • said virus is an adeno-associated virus 8.
  • said virus is an adeno-associated virus 6.
  • said protein is Nme2Cas9.
  • said protein further comprises a nuclear localization signal protein.
  • said nucleotide deaminase is a cytidine deaminase.
  • said nucleotide deaminase is an adenosine deaminase.
  • the protein further comprises a uracil glycosylase inhibitor.
  • the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS.
  • said binding region is a protospacer accessory motif interacting domain.
  • said protospacer accessory motif interacting domain comprises said mutation.
  • said mutation is a D16A mutation.
  • said mutated NmeCas9 protein further comprises CBE4.
  • said mutated NmeCas9 protein further comprises a linker.
  • said linker is a 73aa linker.
  • said linker is a 3xHA-tag.
  • the present invention contemplates a construct, wherein said construct is an optimized nNme2Cas9-ABEmax.
  • the present invention contemplates a construct, wherein said construct is a nNme2Cas9-CBE4.
  • the present invention contemplates a construct, wherein said construct is a YE1-BE3-nNme2Cas9 (D16A)-UGI.
  • the present invention contemplates a method, comprising: a) providing; i) a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N 4 CC nucleotide sequence; ii) a mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N 4 CC nucleotide sequence; b) contacting said nucleotide sequence with said mutated NmeCas9 protein under conditions such that said binding region attaches to said N 4 CC nucleotide sequence; and c) replacing said mutated single base with a wild type base with said mutated NmeCas9 protein.
  • said protein is Nme2Cas9. In one embodiment, said protein further comprises a nuclear localization signal protein. In one embodiment, said nucleotide deaminase is a cytidine deaminase. In one embodiment, said nucleotide deaminase is an adenosine deaminase. In one embodiment, the protein further comprises a uracil glycosylase inhibitor. In one embodiment, the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS. In one embodiment, said binding region is a protospacer accessory motif interacting domain.
  • said protospacer accessory motif interacting domain comprises said mutation.
  • said mutation is a D16A mutation.
  • said mutated NmeCas9 protein further comprises CBE4.
  • said mutated NmeCas9 protein further comprises a linker.
  • said linker is a 73aa linker.
  • said linker is a 3xHA-tag.
  • said gene encodes a tyrosinase.
  • said gene is Fah.
  • said gene is c-fos.
  • the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N 4 CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) an adeno-associated virus comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N 4 CC nucleotide sequence; b) treating said patient with said adeno- associated virus under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
  • said gene encodes a tyrosinase protein.
  • said genetically-based medical condition is tyrosinemia.
  • said virus is an adeno-associated virus 8.
  • said virus is an adeno-associated virus 6.
  • said protein is Nme2Cas9.
  • said protein further comprises a nuclear localization signal protein.
  • said nucleotide deaminase is a cytidine deaminase.
  • said nucleotide deaminase is an adenosine deaminase.
  • the protein further comprises a uracil glycosylase inhibitor.
  • the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS.
  • said binding region is a protospacer accessory motif interacting domain.
  • said protospacer accessory motif interacting domain comprises said mutation.
  • said mutation is a D16A mutation.
  • said mutated NmeCas9 protein further comprises CBE4.
  • said mutated NmeCas9 protein further comprises a linker.
  • said linker is a 73aa linker.
  • said linker is a 3xHA-tag.
  • said gene encodes a tyrosinase.
  • said gene is Fah. In one embodiment, said gene is c-fos. In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N 4 CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) an optimized nNme2Cas9-ABEmax, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N 4 CC nucleotide sequence; b) treating said patient with said optimized nNme2Cas9-ABEmax under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based
  • the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N 4 CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) a nNme2Cas9-CBE4, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N 4 CC nucleotide sequence; b) treating said patient with said nNme2Cas9-CBE4 under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
  • the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N 4 CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) a YE1-BE3-nNme2Cas9 (D16A)-UGI, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N 4 CC nucleotide sequence; b) treating said patient with said nNme2Cas9-CBE4 under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
  • the term“edit”“editing” or“edited” refers to a method of altering a nucleic acid sequence of a polynucleotide (e.g., for example, a wild type naturally occurring nucleic acid sequence or a mutated naturally occurring sequence) by selective deletion of a specific genomic target.
  • a specific genomic target includes, but is not limited to, a chromosomal region, a gene, a promoter, an open reading frame or any nucleic acid sequence.
  • single base refers to one, and only one, nucleotide within a nucleic acid sequence.
  • single base editing it is meant that the base at a specific position within the nucleic acid sequence is replaced with a different base. This replacement may occur by many mechanisms, including but not limited to, substitution or modification.
  • target or“target site” refers to a pre-identified nucleic acid sequence of any composition and/or length.
  • target sites include, but is not limited to, a chromosomal region, a gene, a promoter, an open reading frame or any nucleic acid sequence.
  • the present invention interrogates these specific genomic target sequences with complementary sequences of gRNA.
  • on-target binding sequence refers to a subsequence of a specific genomic target that may be completely complementary to a programmable DNA binding domain and/or a single guide RNA sequence.
  • off-target binding sequence refers to a subsequence of a specific genomic target that may be partially complementary to a programmable DNA binding domain and/or a single guide RNA sequence.
  • compositions comprising a therapeutic agent that achieves a clinically beneficial result (i.e., for example, a reduction of symptoms).
  • Toxicity and therapeutic efficacy of such compositions can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 50 (the dose lethal to 50% of the population) and the ED 50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio LD 50 /ED 50 .
  • Compounds that exhibit large therapeutic indices are preferred.
  • the data obtained from these cell culture assays and additional animal studies can be used in formulating a range of dosage for human use.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
  • symptom refers to any subjective or objective evidence of disease or physical disturbance observed by the patient.
  • subjective evidence is usually based upon patient self-reporting and may include, but is not limited to, pain, headache, visual disturbances, nausea and/or vomiting.
  • objective evidence is usually a result of medical testing including, but not limited to, body temperature, complete blood count, lipid panels, thyroid panels, blood pressure, heart rate, electrocardiogram, tissue and/or body imaging scans.
  • disease or“medical condition”, as used herein, refers to any impairment of the normal state of the living animal or plant body or one of its parts that interrupts or modifies the performance of the vital functions. Typically manifested by distinguishing signs and symptoms, it is usually a response to: i) environmental factors (as malnutrition, industrial hazards, or climate); ii) specific infective agents (as worms, bacteria, or viruses); iii) inherent defects of the organism (as genetic anomalies); and/or iv) combinations of these factors.
  • the quantity and/or magnitude of the symptoms in the treated subject is at least 10% lower than, at least 25% lower than, at least 50% lower than, at least 75% lower than, and/or at least 90% lower than the quantity and/or magnitude of the symptoms in the untreated subject.
  • Attachment refers to any interaction between a medium (or carrier) and a drug. Attachment may be reversible or irreversible. Such attachment includes, but is not limited to, covalent bonding, ionic bonding, Van der Waais forces or friction, and the like.
  • a drug is attached to a medium (or carrier) if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc.
  • drug refers to any pharmacologically active substance capable of being administered which achieves a desired effect.
  • Drugs or compounds can be synthetic or naturally occurring, non-peptide, proteins or peptides, oligonucleotides or nucleotides, polysaccharides or sugars.
  • administering refers to any method of providing a composition to a patient such that the composition has its intended effect on the patient.
  • An exemplary method of administering is by a direct mechanism such as, local tissue administration (i.e., for example, extravascular placement), oral ingestion, transdermal patch, topical, inhalation, suppository etc.
  • patient or“subject”, as used herein, is a human or animal and need not be hospitalized.
  • out-patients persons in nursing homes are "patients.”
  • a patient may comprise any age of a human or non-human animal and therefore includes both adult and juveniles (i.e., children). It is not intended that the term "patient” connote a need for medical treatment, therefore, a patient may voluntarily or involuntarily be part of experimentation whether clinical or in support of basic science studies.
  • affinity refers to any attractive force between substances or particles that causes them to enter into and remain in chemical combination.
  • an inhibitor compound that has a high affinity for a receptor will provide greater efficacy in preventing the receptor from interacting with its natural ligands, than an inhibitor with a low affinity.
  • pharmaceutically or “pharmacologically acceptable”, as used herein, refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
  • pharmaceutically acceptable carrier includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, coatings, isotonic and absorption delaying agents, liposome, commercially available cleansers, and the like. Supplementary bioactive ingredients also can be incorporated into such carriers.
  • viral vector encompasses any nucleic acid construct derived from a virus genome capable of incorporating heterologous nucleic acid sequences for expression in a host organism.
  • viral vectors may include, but are not limited to, adeno-associated viral vectors, lentiviral vectors, SV40 viral vectors, retroviral vectors, adenoviral vectors.
  • viral vectors are occasionally created from pathogenic viruses, they may be modified in such a way as to minimize their overall health risk. This usually involves the deletion of a part of the viral genome involved with viral replication. Such a virus can efficiently infect cells but, once the infection has taken place, the virus may require a helper virus to provide the missing proteins for production of new virions.
  • viral vectors should have a minimal effect on the physiology of the cell it infects and exhibit genetically stable properties (e.g., do not undergo spontaneous genome rearrangement).
  • Most viral vectors are engineered to infect as wide a range of cell types as possible. Even so, a viral receptor can be modified to target the virus to a specific kind of cell. Viruses modified in this manner are said to be pseudotyped.
  • Viral vectors are often engineered to incorporate certain genes that help identify which cells took up the viral genes. These genes are called marker genes. For example, a common marker gene confers antibiotic resistance to a certain antibiotic.
  • the“ROSA26 gene” or“Rosa26 gene” refers to a human or mouse (respectively) locus that is widely used for achieving generalized expression in the mouse.
  • Targeting to the ROSA26 locus may be achieved by introducing a desired gene into the first intron of the locus, at a unique XbaI site approximately 248 bp upstream of the original gene trap line.
  • a construct may be constructed using an adenovirus splice acceptor followed by a gene of interest and a polyadenylation site inserted at the unique XbaI site.
  • a neomycin resistance cassette may also be included in the targeting vector.
  • PCSK9 gene or“Pcsk9 gene” refers to a human or mouse
  • the PCSK9 gene resides on chromosome 1 at the band 1p32.3 and includes 13 exons. This gene may produce at least two isoforms through alternative splicing.
  • proprotein convertase subtilisin/kexin type 9 and“PCSK9” refers to a protein encoded by a gene that modulates low density lipoprotein levels.
  • Proprotein convertase subtilisin/kexin type 9 also known as PCSK9, is an enzyme that in humans is encoded by the PCSK9 gene.
  • Seidah et al. "The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation" Proc. Natl. Acad. Sci. U.S.A.100 (3): 928–933 (2003). Similar genes (orthologs) are found across many species.
  • PSCK9 Many enzymes, including PSCK9, are inactive when they are first synthesized, because they have a section of peptide chains that blocks their activity; proprotein convertases remove that section to activate the enzyme.
  • PSCK9 is believed to play a regulatory role in cholesterol homeostasis.
  • PCSK9 can bind to the epidermal growth factor-like repeat A (EGF-A) domain of the low-density lipoprotein receptor (LDL-R) resulting in LDL-R internalization and degradation.
  • EGF-A epidermal growth factor-like repeat A
  • LDL-R low-density lipoprotein receptor
  • hypercholesterolemia refers to any medical condition wherein blood cholesterol levels are elevated above the clinically recommended levels. For example, if cholesterol is measured using low density lipoproteins (LDLs), hypercholesterolemia may exist if the measured LDL levels are above, for example, approximately 70 mg/dl. Alternatively, if cholesterol is measured using free plasma cholesterol, hypercholesterolemia may exist if the measured free cholesterol levels are above, for example, approximately 200-220 mg/dl.
  • LDLs low density lipoproteins
  • hypercholesterolemia may exist if the measured free cholesterol levels are above, for example, approximately 200-220 mg/dl.
  • CRISPRs As used herein, the term“CRISPRs” or“Clustered Regularly Interspaced Short
  • Palindromic Repeats refers to an acronym for DNA loci that contain multiple, short, direct repetitions of base sequences. Each repetition contains a series of bases followed by 30 or so base pairs known as "spacer DNA".
  • the spacers are short segments of DNA from a virus and may serve as a 'memory' of past exposures to facilitate an adaptive defense against future invasions.
  • CRISPR-associated (cas) refers to genes often associated with CRISPR repeat-spacer arrays.
  • Cas9 refers to a nuclease from Type II CRISPR systems, an enzyme specialized for generating double-strand breaks in DNA, with two active cutting sites (the HNH and RuvC domains), one for each strand of the double helix.
  • Jinek combined tracrRNA and spacer RNA into a "single-guide RNA" (sgRNA) molecule that, mixed with Cas9, could find and cleave DNA targets through Watson-Crick pairing between the guide sequence within the sgRNA and the target DNA sequence.
  • sgRNA single-guide RNA
  • PAM protospacer adjacent motif
  • Cas9/sgRNA DNA sequence that may be required for a Cas9/sgRNA to form an R-loop to interrogate a specific DNA sequence through Watson-Crick pairing of its guide RNA with the genome.
  • the PAM specificity may be a function of the DNA-binding specificity of the Cas9 protein (e.g., a “protospacer adjacent motif recognition domain” at the C-terminus of Cas9).
  • sgRNA refers to single guide RNA used in conjunction with CRISPR associated systems (Cas). sgRNAs are a fusion of crRNA and tracrRNA and contain nucleotides of sequence complementary to the desired target site.
  • fluorescent protein refers to a protein domain that comprises at least one organic compound moiety that emits fluorescent light in response to the appropriate wavelengths.
  • fluorescent proteins may emit red, blue and/or green light.
  • Such proteins are readily commercially available including, but not limited to: i) mCherry (Clonetech Laboratories): excitation: 556/20 nm (wavelength/bandwidth); emission: 630/91 nm; ii) sfGFP (Invitrogen): excitation: 470/28 nm; emission: 512/23 nm; iii) TagBFP (Evrogen): excitation 387/11 nm; emission 464/23 nm.
  • sgRNA refers to single guide RNA used in conjunction with CRISPR associated systems (Cas). sgRNAs contains nucleotides of sequence complementary to the desired target site. Watson-crick pairing of the sgRNA with the target site recruits the nuclease-deficient Cas9 to bind the DNA at that locus.
  • orthogonal refers targets that are non-overlapping, uncorrelated, or independent. For example, if two orthogonal nuclease-deficient Cas9 gene fused to different effector domains were implemented, the sgRNAs coded for each would not cross- talk or overlap. Not all nuclease-deficient Cas9 genes operate the same, which enables the use of orthogonal nuclease-deficient Cas9 gene fused to a different effector domains provided the appropriate orthogonal sgRNAs.
  • phenotypic change or“phenotype” refers to the composite of an organism's observable characteristics or traits, such as its morphology, development, biochemical or physiological properties, phenology, behavior, and products of behavior.
  • Phenotypes result from the expression of an organism's genes as well as the influence of environmental factors and the interactions between the two.
  • nucleic acid sequence and “nucleotide sequence” as used herein refer to an
  • an isolated nucleic acid refers to any nucleic acid molecule that has been removed from its natural state (e.g., removed from a cell and is, in a preferred embodiment, free of other genomic nucleic acid).
  • amino acid sequence and “polypeptide sequence” as used herein, are interchangeable and to refer to a sequence of amino acids.
  • portion when in reference to a protein (as in “a portion of a given protein”) refers to fragments of that protein.
  • the fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.
  • portion when used in reference to a nucleotide sequence refers to fragments of that nucleotide sequence.
  • the fragments may range in size from 5 nucleotide residues to the entire nucleotide sequence minus one nucleic acid residue.
  • the terms “complementary” or “complementarity” are used in reference to “polynucleotides” and “oligonucleotides” (which are interchangeable terms that refer to a sequence of nucleotides) related by the base-pairing rules.
  • the sequence "C-A-G- T,” is complementary to the sequence "G-T-C-A.”
  • Complementarity can be “partial” or “total.”
  • Partial complementarity is where one or more nucleic acid bases is not matched according to the base pairing rules.
  • Total or “complete” complementarity between nucleic acids is where each and every nucleic acid base is matched with another base under the base pairing rules.
  • the degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.
  • nucleotide sequences refer to a degree of complementarity with other nucleotide sequences. There may be partial homology or complete homology (i.e., identity).
  • a nucleotide sequence which is partially complementary, i.e., “substantially homologous,” to a nucleic acid sequence is one that at least partially inhibits a completely complementary sequence from hybridizing to a target nucleic acid sequence. The inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or Northern blot, solution hybridization and the like) under conditions of low stringency.
  • a substantially homologous sequence or probe will compete for and inhibit the binding (i.e., the hybridization) of a completely homologous sequence to a target sequence under conditions of low stringency. This is not to say that conditions of low stringency are such that non-specific binding is permitted; low stringency conditions require that the binding of two sequences to one another be a specific (i.e., selective) interaction.
  • the absence of non-specific binding may be tested by the use of a second target sequence which lacks even a partial degree of complementarity (e.g., less than about 30% identity); in the absence of non-specific binding the probe will not hybridize to the second non-complementary target.
  • amino acid sequences refer to the degree of identity of the primary structure between two amino acid sequences. Such a degree of identity may be directed to a portion of each amino acid sequence, or to the entire length of the amino acid sequence.
  • Two or more amino acid sequences that are “substantially homologous” may have at least 50% identity, preferably at least 75% identity, more preferably at least 85% identity, most preferably at least 95%, or 100% identity.
  • oligonucleotide sequence which is a "homolog" is defined herein as an
  • oligonucleotide sequence which exhibits greater than or equal to 50% identity to a sequence, when sequences having a length of 100 bp or larger are compared.
  • Low stringency conditions comprise conditions equivalent to binding or hybridization at 42°C in a solution consisting of 5 x SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 ⁇ H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5x Denhardt's reagent ⁇ 50x Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; Sigma) ⁇ and 100 mg/ml denatured salmon sperm DNA followed by washing in a solution comprising 5x SSPE, 0.1% SDS at 42°C when a probe of about 500 nucleotides in length is employed.
  • 5 x SSPE 43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 ⁇ H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH
  • low stringency conditions may also be employed to comprise low stringency conditions; factors such as the length and nature (DNA, RNA, base composition) of the probe and nature of the target ( DNA, RNA, base composition, present in solution or immobilized, etc.) and the concentration of the salts and other components (e.g., the presence or absence of formamide, dextran sulfate, polyethylene glycol), as well as components of the hybridization solution may be varied to generate conditions of low stringency hybridization different from, but equivalent to, the above listed conditions.
  • conditions which promote hybridization under conditions of high stringency e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc.
  • high stringency e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc.
  • hybridization is used in reference to the pairing of
  • hybridization and the strength of hybridization is impacted by such factors as the degree of complementarity between the nucleic acids, stringency of the conditions involved, the T m of the formed hybrid, and the G:C ratio within the nucleic acids.
  • hybridization complex refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bounds between complementary G and C bases and between complementary A and T bases; these hydrogen bonds may be further stabilized by base stacking interactions.
  • the two complementary nucleic acid sequences hydrogen bond in an antiparallel configuration.
  • a hybridization complex may be formed in solution (e.g., C 0 t or R 0 t analysis) or between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized to a solid support (e.g., a nylon membrane or a nitrocellulose filter as employed in Southern and Northern blotting, dot blotting or a glass slide as employed in in situ hybridization, including FISH (fluorescent in situ hybridization)).
  • a solid support e.g., a nylon membrane or a nitrocellulose filter as employed in Southern and Northern blotting, dot blotting or a glass slide as employed in in situ hybridization, including FISH (fluorescent in situ hybridization)
  • DNA molecules are said to have "5' ends” and "3' ends” because mononucleotides are reacted to make oligonucleotides in a manner such that the 5' phosphate of one mononucleotide pentose ring is attached to the 3' oxygen of its neighbor in one direction via a phosphodiester linkage. Therefore, an end of an oligonucleotide is referred to as the "5' end” if its 5' phosphate is not linked to the 3' oxygen of a mononucleotide pentose ring. An end of an oligonucleotide is referred to as the "3' end” if its 3' oxygen is not linked to a 5' phosphate of another
  • a nucleic acid sequence even if internal to a larger oligonucleotide, also may be said to have 5' and 3' ends.
  • discrete elements are referred to as being "upstream” or 5' of the "downstream” or 3' elements. This terminology reflects the fact that transcription proceeds in a 5' to 3' fashion along the DNA strand.
  • the promoter and enhancer elements which direct transcription of a linked gene are generally located 5' or upstream of the coding region. However, enhancer elements can exert their effect even when located 3' of the promoter element and the coding region. Transcription termination and polyadenylation signals are located 3' or downstream of the coding region.
  • transfection or "transfected” refers to the introduction of foreign DNA into a cell.
  • nucleic acid molecule encoding refers to the order or sequence of deoxyribonucleotides along a strand of deoxyribonucleic acid. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide (protein) chain. The DNA sequence thus codes for the amino acid sequence.
  • the term “gene” means the deoxyribonucleotide sequences comprising the coding region of a structural gene and including sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of about 1 kb on either end such that the gene corresponds to the length of the full-length mRNA.
  • the sequences which are located 5' of the coding region and which are present on the mRNA are referred to as 5' non-translated sequences.
  • the sequences which are located 3' or downstream of the coding region and which are present on the mRNA are referred to as 3' non-translated sequences.
  • the term “gene” encompasses both cDNA and genomic forms of a gene.
  • a genomic form or clone of a gene contains the coding region interrupted with non-coding sequences termed "introns" or "intervening regions” or “intervening sequences.” Introns are segments of a gene which are transcribed into
  • heterogeneous nuclear RNA introns may contain regulatory elements such as enhancers. Introns are removed or "spliced out” from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (mRNA) transcript.
  • mRNA messenger RNA
  • genomic forms of a gene may also include sequences located on both the 5' and 3' end of the sequences which are present on the RNA transcript.
  • flanking sequences or regions are located 5' or 3' to the non-translated sequences present on the mRNA transcript.
  • the 5' flanking region may contain regulatory sequences such as promoters and enhancers which control or influence the transcription of the gene.
  • the 3' flanking region may contain sequences which direct the termination of transcription, posttranscriptional cleavage and polyadenylation.
  • label or “detectable label” are used herein, to refer to any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means.
  • labels include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., Dynabeads ® ), fluorescent dyes (e.g., fluorescein, texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and calorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein, texas red, rhodamine, green fluorescent protein,
  • Patents teaching the use of such labels include, but are not limited to, U.S. Pat. Nos.3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241 (all herein incorporated by reference in their entirety).
  • the labels contemplated in the present invention may be detected by many methods. For example, radiolabels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted light.
  • Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting, the reaction product produced by the action of the enzyme on the substrate, and calorimetric labels are detected by simply visualizing the colored label.
  • Figure 1 illustrates exemplary schematic embodiments of an NmeCas9 deaminase fusion protein single base editor and exemplary constructed plasmids of base editors.
  • Figure 1A shows an exemplary YE1-BE3-nNme2Cas9 (D16A)-UGI construct.
  • Figure 1B shows an exemplary ABE7.10 nNme2Cas9 (D16A) construct.
  • Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct comprising two SV40 NLS sequences.
  • Figure 1D shows an exemplary nNme2Cas9-CBE4 (also called a BE4-nNme2Cas9
  • Figure 1E shows an exemplary optimized nNme2Cas9-ABEmax construct.
  • Figure 2 presents exemplary data of the electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3-nNme2Cas9 (D16A)-UGI fusion protein efficiently converting C to T at endogenous target site 25 (TS25) in HEK293T cells via nucleofection.
  • D16A YE1-BE3-nNme2Cas9
  • Figure 2A shows exemplary sequences for a TS25 endogenous target site (within the black rectangle). GN23 sgRNA base-pairs with the target DNA strand, leaving the displaced DNA strand for cytidine deaminase to edit (e.g. new green nucleotides).
  • Figure 2B shows exemplary sequencing data showing a doublet nucleotide peak (7 th position from 5’ end; arrow) demonstrating the successful single base editing of a cytidine to a thymidine (e.g., a C•G base pair conversion to a T•A base pair).
  • Figure 2C shows an exemplary quantitation of the data shown in Figure 2B plotting the percent conversion of C ® T single base editing.
  • The“no sgRNA” control displays the background noise due to Sanger sequencing. EditR (Kluesner et al., 2018) was used to perform the analysis.
  • Figure 3 presents exemplary specific UGI target sites that were respectively integrated into YE1-BE3-nNme2Cas9/D16A mutant fusion proteins and co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line. Converted bases are highlighted in orange color. Background signals were filtered using negative control samples (YE1-BE3- nNme2Cas9 nucleofected K562 cells without sgRNA constructs). N 4 CC PAMs are boxed. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column.
  • EGFP enhanced green fluorescent protein
  • Figure 3A shows an exemplary EGFP-Site 1.
  • Figure 3B shows an exemplary EGFP-Site 2.
  • Figure 3C shows an exemplary EGFP-Site 3.
  • Figure 3D shows an exemplary EGFP-Site 4.
  • Figure 3E shows an exemplary deep-sequencing analysis indicating where YE1-BE3- nNme2Cas9 converts C residues to T residues at endogenous c-fos promoter region.
  • the percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column.
  • the converted bases are highlighted in orange or yellow color. Background signals were filtered using negative control samples. The highest percentage of editing is 32.50%.
  • Figure 3F shows an exemplary deep-sequencing analysis indicating where ABE7.10- nNme2Cas9 or ABEmax (Koblan et al., 2018)-nNme2Cas9 converts A residues to G residues at endogenous c-fos promoter region.
  • the percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange color. Background signals were filtered using negative control samples.
  • the percentage of editing is 0.53% by ABE7.10-nNme2Cas9 or 2.33% by ABEmax-nNme2Cas9 (D16A).
  • Figure 4 presents an exemplary alignment of the wildtype Fah gene with the tyrosinemia Fah mutant gene showing an A-G single base gene editing target site (position 9).
  • the respective SpyCas9 single PAM site and NmeCas9 double PAM sites are indicated for demonstrating the suboptimal targeting window relative to the SpyCas9 PAM site.
  • Figure 5 illustrates exemplary three closely related Neisseria meningitidis Cas9 orthologs that have distinct PAMs.
  • Figure 5A shows an exemplary schematic showing mutated residues (orange spheres) between Nme2Cas9 (left) and Nme3Cas9 (right) mapped onto the predicted structure of Nme1Cas9, revealing the cluster of mutations in the PID (black).
  • Figure 5B shows an exemplary experimental workflow of the in vitro PAM discovery assay with a 10-bp randomized PAM region. Following in vitro digestion, adapters were ligated to cleaved products for library construction and sequencing.
  • Figure 5C shows exemplary sequence logos resulting from in vitro PAM discovery reveal the enrichment of a N 4 GATT PAM for Nme1Cas9, consistent with its previously established specificity.
  • Figure 5D shows exemplary sequence logos indicating that Nme1Cas9 with its PID
  • FIG. 6 shows an exemplary sequence logo showing that full-length Nme2Cas9 recognizes an N 4 CC PAM, based on efficient substrate cleavage of a target pool with a fixed C at PAM position 5, and with PAM nts 1-4 and 6-8 randomized.
  • Figure 6 presents a characterization of Neisseria meningitidis Cas9 orthologs with rapidly-evolving PIDs, as related to Figure 5.
  • Figure 6A shows an exemplary unrooted phylogenetic tree of NmeCas9 orthologs that are >80% identical to Nme1Cas9. Three distinct branches emerged, with the majority of mutations clustered in the PID. Groups 1 (blue), 2 (orange), and 3 (green) have PIDs with >98%, ⁇ 52%, and ⁇ 86% identity to Nme1Cas9, respectively. Three representative Cas9 orthologs (one from each group)
  • Figure 6B shows an exemplary schematic showing the CRISPR-cas loci of the strains encoding the three Cas9 orthologs (Nme1Cas9, Nme2Cas9, and Nme3Cas9) from (A). Percent identities of each CRISPR-Cas component with N. meningitidis 8013 (encoding Nme1Cas9) are shown. Blue and red arrows denote pre-crRNA and tracrRNA transcription initiation sites, respectively.
  • Figure 6C shows an exemplary normalized read counts (% of total reads) from cleaved DNAs from the in vitro assays for intact Nme1Cas9 (grey), for chimeras with Nme1Cas9’s PID swapped with those of Nme2Cas9 and Nme3Cas9 (mixed colors), and for full-length Nme2Cas9 (orange), are plotted.
  • the reduced normalized read counts indicate lower cleavage efficiencies in the chimeras.
  • Figure 6D shows an exemplary sequence logos from the in vitro PAM discovery assay on an NNNNCNNN PAM pool by Nme1Cas9 with its PID swapped with those of Nme2Cas9 (left) or Nme3Cas9 (right).
  • Figure 7 presents exemplary data showing that Nme2Cas9 uses a 22-24 nt spacer to edit sites adjacent to an N 4 CC PAM. All experiments were done in triplicate, and error bars represent the standard error of the mean (s.e.m.).
  • Figure 7A shows an exemplary schematic diagram depicting transient transfection and editing of HEK293T TLR2.0 cells, with mCherry+ cells detected by flow cytometry 72 hours after transfection.
  • Figure 7B shows an exemplary Nme2Cas9 editing of the TLR2.0 reporter. Sites with N 4 CC PAMs were targeted with varying efficiencies, while no Nme2Cas9 targeting was observed at an N 4 GATT PAM or in the absence of sgRNA.
  • SpyCas9 targeting a previously validated site with an NGG PAM
  • Nme1Cas9 (targeting N 4 GATT) were used as positive controls.
  • Figure 7C shows an exemplary effect of spacer length on the efficiency of Nme2Cas9 editing.
  • Figure 7D shows an exemplary An Nme2Cas9 dual nickase can be used in tandem to generate NHEJ- and HDR-based edits in TLR2.0.
  • HNH nickase HNH nickase
  • Nme2Cas9 D16A Nme2Cas9 D16A ; RuvC nickase, Nme2Cas9 H588A .
  • Cleavage sites 32 bp and 64 bp apart were targeted using either nickase.
  • the HNH nickase (Nme2Cas9 D16A ) yielded efficient editing, particularly with the cleavage sites that were separated by 32 bp, whereas the RuvC nickase (Nme2Cas9 H588A ) was not effective.
  • Wildtype Nme2Cas9 was used as a control.
  • Figure 8 presents exemplary data showing PAM, spacer, and seed requirements for Nme2Cas9 targeting in mammalian cells, as related to Figure 7. All experiments were done in triplicate and error bars represent s.e.m.
  • Figure 8A shows an exemplary Nme2Cas9 targeting at N 4 CD sites in TLR2.0, with
  • Figure 8B shows an exemplary Nme2Cas9 targeting at N 4 DC sites in TLR2.0 [similar to (A)].
  • Figure 8C shows exemplary guide truncations on a TLR2.0 site (distinct from that in Figure 2C) with a N 4 CCA PAM, revealing similar length requirements as those observed at the other site.
  • Figure 8D shows exemplary Nme2Cas9 targeting efficiency is differentially sensitive to single-nucleotide mismatches in the seed region of the sgRNA. Data show the effects of walking single-nucleotide sgRNA mismatches along the 23-nt spacer in a TLR2.0 target site.
  • Figure 9 presents exemplary data showing Nme2Cas9 genome editing at endogenous loci in mammalian cells via multiple delivery methods. All results represent 3 independent biological replicates, and error bars represent s.e.m.
  • Figure 9A shows an exemplary Nme2Cas9 genome editing of endogenous human sites in HEK293T cells following transient transfection of Nme2Cas9- and sgRNA- expressing plasmids.40 sites were screened initially (Table 1); the 14 sites shown (selected to include representatives of varying editing efficiencies, as measured by TIDE) were then re-analyzed in triplicate. An Nme1Cas9 target site (with an N 4 GATT PAM) was used as a negative control.
  • Figure 9B shows exemplary data charts: Left panel: Transient transfection of a single plasmid expressing both Nme2Cas9 and sgRNA (targeting the Pcsk9 and Rosa26 loci) enables editing in Hepa1-6 mouse cells, as detected by TIDE. Right panel: Electroporation of sgRNA plasmids into K562 cells stably expressing Nme2Cas9 from a lentivector results in efficient indel formation.
  • Figure 9C shows exemplary Nme2Cas9 can be electroporated as an RNP complex to induce genome editing.40 picomoles Cas9 along with 50 picomoles of in vitro- transcribed sgRNAs targeting three different loci were electroporated into HEK293T cells. Indels were measured after 72h using TIDE.
  • Figure 10 presents exemplary data showing dose dependence and segmental deletions by Nme2Cas9, as related to Figure 9.
  • Figure 10A shows exemplary increasing the dose of electroporated Nme2Cas9 plasmid (500 ng, vs.200 ng in Figure 3A) improves editing efficiency at two sites (TS16 and TS6). Data provided in yellow are re-used from Figure 9A.
  • Figure 10B shows exemplary Nme2Cas9 can be used to create precise segmental
  • FIG. 11 presents exemplary data showing that Nme2Cas9 is subject to inhibition by a subset of type II-C anti-CRISPR families in vitro and in cells. All experiments were done in triplicate and error bars represent s.e.m.
  • Figure 11A shows exemplary In vitro cleavage assay of Nme1Cas9 and Nme2Cas9 in the presence of five previously characterized anti-CRISPR proteins (10:1 ratio of Acr:Cas9).
  • Figure 11B shows exemplary genome editing in the presence of the five previously
  • Figure 11C shows exemplary Acr inhibition of Nme2Cas9 is dose-dependent with
  • Nme2Cas9 is fully inhibited by AcrIIC1 Nme and AcrIIC4 Hpa at 2:1 and 1:1 mass ratios of cotransfected Acr and Nme2Cas9 plasmids, respectively.
  • Figure 12 presents exemplary data showing that a Nme2Cas9 PID swap renders
  • Nme1Cas9 insensitive to AcrIIC5 Smu inhibition as related to Figure 11.
  • Nme1Cas9-Nme2Cas9PID chimera in the presence of previously characterized Acr proteins (10 uM Cas9-sgRNA + 100 uM Acr).
  • Figure 13 presents exemplary data showing orthogonality and relative accuracy of Nme2Cas9 and SpyCas9 at dual target sites, as related to Figure 12.
  • Figure 13A shows exemplary Nme2Cas9 and SpyCas9 guides are orthogonal.
  • Figure 13B shows exemplary Nme2Cas9 and SpyCas9 exhibiting comparable on-target editing efficiencies as assessed by GUIDE-seq. Bars indicate on-target read counts from GUIDE-Seq at the three dual sites targeted by each ortholog. Orange bars represent Nme2Cas9 and black bars represent SpyCas9.
  • Figure 13C shows an exemplary SpyCas9’s on-target vs. off-target read counts for each site. Orange bars represent the on-target reads while black bars represent off- targets.
  • Figure 13D shows exemplary Nme2Cas9’s on-target vs. off-target reads for each site.
  • Figure 13E bar graphs showing exemplary indel efficiencies (measured by TIDE) at potential off-target sites predicted by CRISPRSeek. On- and off-target site sequences are shown on the left, with the PAM region underlined and sgRNA mismatches and non-consensus PAM nucleotides given in red.
  • Figure 14 presents exemplary data showing that Nme2Cas9 exhibits little or no detectable off-targeting in mammalian cells.
  • Figure 14A shows an exemplary schematic depicting dual sites (DSs) targetable by both SpyCas9 and Nme2Cas9 by virtue of their non-overlapping PAMs.
  • Nme2Cas9 PAM (orange) and SpyCas9 PAM (blue) are highlighted.
  • a 24nt Nme2Cas9 guide sequence is indicated in yellow; the corresponding guide sequence for SpyCas9 would be 4nt shorter at the 5’ end.
  • Figure 14B shows an exemplary Nme2Cas9 and SpyCas9 that both induce indels at DSs.
  • FIG. 14C shows exemplary Nme2Cas9 genome editing that is highly accurate in human cells. Numbers of off-target sites detected by GUIDE-Seq for each nuclease at individual target sites are shown. In addition to dual sites, we analyzed TS6 (because of its high on-target editing efficiency) and Pcsk9 and Rosa26 sites in mouse Hepa1-6 cells (to measure accuracy in another cell type).
  • Figure 14D shows an exemplary targeted deep sequencing to detect indels in edited cells confirms the high Nme2Cas9 accuracy indicated by GUIDE-seq.
  • Figure 14E shows an exemplary sequence for the validated off-target site of the Rosa26 guide, showing the PAM region (underlined), the consensus CC PAM dinucleotide (bold), and three mismatches in the PAM-distal portion of the spacer (red).
  • Figure 15 presents exemplary data showing Nme2Cas9 genome editing in vivo via all-in- one AAV delivery.
  • Figure 15A shows exemplary workflow for delivery of AAV8.sgRNA.Nme2Cas9 to lower cholesterol levels in mice by targeting Pcsk9.
  • Top schematic of the all-in- one AAV vector expressing Nme2Cas9 and the sgRNA (individual genome elements not to scale).
  • BGH bovine growth hormone poly(A) site;
  • HA epitope tag;
  • NLS nuclear localization sequence;
  • h human-codon-optimized.
  • Bottom Timeline for AAV8.sgRNA.Nme2Cas9 tail-vein injections (4 x 10 11 GCs), followed by cholesterol measurements at day 14 and indel, histology and cholesterol analyses at day 28 post-injection.
  • Figure 15B shows an exemplary TIDE analysis to measure indels in DNA extracted from livers of mice injected with AAV8.Nme2Cas9+sgRNA targeting Pcsk9 and Rosa26 (control) loci. Indel efficiency at the lone off-target site identified by GUIDE-seq for these two sgRNAs (Rosa26
  • Figure 15C shows an exemplary reduced serum cholesterol levels in mice injected with the Pcsk9-targeting guide compared to the Rosa26-targeting controls. P values are calculated by unpaired two-tailed t-test.
  • Figure 16 presents exemplary data showing PCSK9 knockdown and liver histology following Nme2Cas9 AAV delivery and editing, related to Figure 15.
  • Figure 16A shows exemplary Western blotting using anti-PCSK9 antibody reveals strongly reduced levels of PCSK9 in the livers of mice treated with sgPcsk9, compared to mice treated with sgRosa26.2ng of recombinant PCSK9 was used as a mobility standard (left-most lane), and a cross-reacting band in the liver samples is indicated by an asterisk. GAPDH was used as loading control (bottom panel).
  • Figure 16B shows exemplary H&E staining from livers of mice injected with
  • Figure 17 presents exemplary data showing Tyr editing ex vivo in mouse zygotes, related to Figure 16.
  • Figure 17A shows an exemplary two sites in Tyr, each with N 4 CC PAMs, were tested for editing in Hepa1-6 cells.
  • the sgTyr2 guide exhibited higher editing efficiency and was selected for further testing.
  • Figure 17B shows an exemplary seven mice that survived post-natal development, and each exhibited coat color phenotypes as well as on-target editing, as assayed by TIDE.
  • Figure 17C shows an exemplary Indel spectra from tail DNA of each mouse from (B), as well as an unedited C57BL/6NJ mouse, as indicated by TIDE analysis.
  • Figure 18 presents exemplary data showing Nme2Cas9 genome editing ex vivo via all-in- one AAV delivery.
  • Figure 18A shows an exemplary workflow for single-AAV Nme2Cas9 editing ex vivo to generate albino C57BL/6NJ mice by targeting the Tyr gene.
  • Zygotes are cultured in KSOM containing AAV6.Nme2Cas9:sgTyr for 5-6 hours, rinsed in M2, and cultured for a day before being transferred to the oviduct of pseudo-pregnant recipients.
  • Figure 18B shows exemplary albino (left) and chinchilla or variegated (middle) mice generated by 3x10 9 GCs, and chinchilla or variegated mice (right) generated by 3x10 8 GCs of zygotes with AAV6.Nme2Cas9:sgTyr.
  • Figure 18C shows an exemplary summary of Nme2Cas9.sgTyr single-AAV ex vivo Tyr editing experiments at two AAV doses.
  • Figure 19 shows an exemplary mCherry reporter assay for nSpCas9-ABEmax and optimized ABEmax-nNme2Cas9 (D16A) activities.
  • Figure 19A shows exemplary sequence information of sequence information of ABE- mCherry reporter.
  • the mCherry signal will show up if the nSpCas9-ABEmax or optimized ABEmax-nNme2Cas9 (D16A) can convert TAG to CAG (which is encoded Gln).
  • Figure 19B shows an exemplary mCherry signals light up since SpCas9-ABE or
  • ABEmax-nNme2Cas9 (D16A) is active in the specific region of the mCherry reporter.
  • Upper panel is the negative control
  • middle panel shows the mCherry signals light up in reporter cells treated with nSpCas9-ABEmax
  • bottom panel shows the mCherry signals light up in reporter cells treated with optimized ABEmax-nNme2Cas9 (D16A).
  • Figure 19C shows an exemplary FACs Quantitation of base editing events in mCherry reporter cells transfected with the SpCas9-ABE or ABEmax-nNme2Cas9 (D16A).
  • Figure 20 shows an exemplary GFP reporter assay for nSpCas9-CBE4 (Addgene
  • Figure 20A shows exemplary sequence information of CBE-GFP reporter.
  • the GFP signal will show up if the nSpCas9- CBE4 or CBE4-nNme2Cas9 (D16A)-UGI-UGI can convert CAC to TAC/TAT
  • Figure 20B shows an exemplary GFP signal (green) since nSpCas9-CBE4 or CBE4- nNme2Cas9 (D16A)-UGI-UGI is active in the specific region of the GFP reporter.
  • Upper panel is the negative control.
  • Middle panel shows that the mCherry signals light up in the reporter cells treated with CBE4-nNme2Cas9 (D16A)-UGI-UGI.
  • Bottom panel shows that the GFP signals light up in the reporter cells treated with CBE4-nNme2Cas9 (D16A)-UGI-UGI).
  • Figure 20C shows an exemplary FACs Quantitation of base editing events in GFP reporter cells transfected with nSpCas9-CBE4 or CBE4-nNme2Cas9 (D16A)-UGI-UGI.
  • N 6; error bars represent S.D. Results are from biological replicates performed in technical duplicates.
  • Figure 21 shows exemplary cytosine editing by CBE4-nNme2Cas9 (D16A)-UGI-UGI.
  • Upper panel shows the KANK3 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples.
  • Bottom panel shows the quantification of the substitution rate of each type of base in the CBE4- nNme2Cas9 (D16A)-UGI-UGI editing window of the KANK3 target sequences.
  • Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red.
  • Figure 22 shows exemplary cytosine and adenine editing by CBE4-nNme2Cas9 (D16A)- UGI-UGI and optimized ABEmax-nNme2Cas9 (D16A), respectively.
  • Upper panel shows the PLXNB2 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples.
  • Middle panel shows the quantification of the substitution rate of each type of base in the optimized ABEmax-nNme2Cas9 (D16A) editing windows of the PLXNB2 target sequences.
  • Sequence tables show nucleotide frequencies at each position. Frequencies of expected A-to-G conversion are highlighted in red.
  • Bottom panel shows the quantification of the substitution rate of each type of base in the CBE4-nNme2Cas9 (D16A)- UGI-UGI editing windows of the PLXNB2 target sequences. Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red.
  • the present invention is related to the field of gene editing.
  • the gene editing is directed toward single nucleotide base editing.
  • single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair.
  • the high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein.
  • the compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein can edit sites that are not targetable by conventional SpyCas9 base editor platforms.
  • Cas9 is a programmable nuclease that uses a guide RNA to create a double-stranded break at any desired genomic locus. This programmability has been harnessed for biomedical and therapeutic approaches. However, Cas9-induced breaks often lead to imprecise repair by the cellular machinery, hindering its therapeutic application for single-base corrections as well as uniform and precise gene knock-outs. Moreover, it is extremely challenging to combine Cas9- induced DNA double strand breaks and a repair template for homologous directed repair (HDR) for correcting genetic mutations in post-mitotic cells (e.g. neuronal cells).
  • HDR homologous directed repair
  • Single nucleotide base editing is a genome editing approach where a nuclease-dead or - impaired Cas9 (e.g., dead Cas9 (dCas9) or nickase Cas9 (nCas9)) is fused to another enzyme capable of base-editing nucleotides without causing DNA double strand breaks.
  • Cas9 base editors Two broad classes of Cas9 base editors have been developed: i) cytidine deaminase (edits a C•G base pair to a T•A base pair) SpyCas9 fusion protein; and ii) adenosine deaminase (edits a A•T base pair to a G•C base pair) SpyCas9.
  • SpyCas9 base editing platforms cannot be used to target all single-base mutations due to their limited editing windows.
  • the editing window is constrained by the requirement for an NGG PAM.
  • SpyCas9 is also intrinsically associated with high off-targeting effects in genome editing.
  • the present invention contemplates a deaminase fusion protein with a compact and hyper-accurate Nme2Cas9 (Neisseria meningitidis spp.).
  • This Nme2Cas9 has 1,082 amino acids as compared to SpyCas9 that has 1,368 amino acids.
  • This Nme2Cas9 ortholog functions efficiently in mammalian cells, recognizes an N 4 CC PAM, and is intrinsically hyper- accurate. Edraki et al., Mol Cell. (in preparation).
  • the present invention contemplates a fusion protein comprising a Nme2Cas9 and a deaminase protein, exemplary examples including ABE7.10-nNme2Cas9 (D16A); Optimized nNme2Cas9-ABEmax; nNme2Cas9-CBE4 (equals BE4-nNme2Cas9 (D16A)-UGI-UGI ) as well as ABEmax-nNme2Cas9 (D16A). See, Figure 1A, Figure 1B, Figure 1C, Figure 1D and Figure 1E.
  • Figure 1 illustrates exemplary schematic embodiments of an NmeCas9 deaminase fusion protein single base editor and exemplary constructed plasmids of base editors.
  • Figure 1A shows an exemplary YE1-BE3-nNme2Cas9 (D16A)-UGI construct.
  • Figure 1B shows an exemplary ABE7.10 nNme2Cas9 (D16A) construct.
  • Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct.
  • Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct comprising two SV40 NLS sequences.
  • Figure 1D shows an exemplary nNme2Cas9-CBE4 (also called a BE4-nNme2Cas9 (D16A)-UGI-UGI) construct.
  • Figure 1E shows an exemplary optimized nNme2Cas9-ABEmax construct.
  • the deaminase protein is Apobec1 (YE1-BE3). It is not intended to limit Apobec1 to one organism.
  • the Apobec1 is derived from a rat species. Kim et al.,“Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions”. Nature Biotechnology 35 (2017).
  • the Nme2Cas9 comprises an nNme2Cas9 D16A mutant.
  • the fusion protein further comprises a uracil glycosylase inhibitor protein (UGI).
  • the fusion protein comprises a YE1-BE3-nNme2Cas9 (D16A)-UGI construct.
  • the YE1-BE3-nNme2Cas9 (D16A)-UGI construct has the sequence of:
  • the present invention contemplates a fusion protein comprising an NmeCas9/ABE7.10 deaminase protein.
  • the deaminase protein is TadA.
  • the deaminase protein is TadA 7.10.
  • the ABE7.10- nNme2Cas9 (D16A) construct has the following sequence:
  • an ABE7.10-nNme2Cas9 (D16A) construct has the following amino acid sequence:
  • an ABEmax-nNme2Cas9 (D16A) construct has the following amino acid sequence:
  • a CBE4-nNme2Cas9 (D16A)-UGI-UGI construct has the following amino acid sequence:
  • an optimized nNme2Cas9-ABEmax construct refers to an optimized version with improved promoter, NLS sequences, and linker sequences.
  • an optimized nNme2Cas9-ABEmax construct comprises, 5' to 3', a C-myc NLS, 12aa linker, 15aa linker, SV40 NLS, TadA, TadA*7.10, 48aa linker, nNme2Cas9, a 73aa linker (3xHA-tag), 15aa linker, and a C-myc NLS.
  • an optimized nNme2Cas9-ABEmax construct further comprises at least two each alternating C-myc NLS and a 12aa linker at the 3' end.
  • an optimized nNme2Cas9-ABEmax construct further comprises at least two each alternating 15aa linker and C-myc NLS at the 5' end. See, Figure 1E for example.
  • an optimized nNme2Cas9-ABEmax construct has the following amino acid sequence:
  • hTadA7.10 underlined
  • hTadA*7.10 underlined/bold
  • linker bold italics
  • nNme2Cas9 italics
  • Cmyc-NLS plain
  • SV40-NLS symbold
  • a plasmid nSpCas9-ABEmax (Addgene ID:112095) was used for experimental controls and for molecular cloning.
  • a plasmid nSpCas9- CBE4 (Addgene ID: 100802) was used for experimental controls and for molecular cloning.
  • Electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3- Nme2Cas9 nucleotide deaminase fusion protein achieved robust single-base editing of a C•G base pair to a T•A base pair at an endogenous target site (TS25). See, Figures 2A-C.
  • Figure 2 presents exemplary data of the electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3-nNme2Cas9 (D16A)-UGI fusion protein efficiently converting C to T at endogenous target site 25 (TS25) in HEK293T cells via nucleofection.
  • Figure 2A shows exemplary sequences for a TS25 endogenous target site (within the black rectangle).
  • GN23 sgRNA base-pairs with the target DNA strand, leaving the displaced DNA strand for cytidine deaminase to edit (e.g. new green nucleotides).
  • Figure 2B shows exemplary sequencing data showing a doublet nucleotide peak (7 th position from 5’ end; arrow) demonstrating the successful single base editing of a cytidine to a thymidine (e.g., a C•G base pair conversion to a T•A base pair).
  • YE1-BE3-nNme2Cas9/D16A mutant fusion proteins were co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line expressing enhanced green fluorescent protein (EGFP).
  • EGFP enhanced green fluorescent protein
  • Each YE1-BE3-nNme2Cas9/D16A mutant fusion protein had a specific UGI target site. See, Figures 3A-D.
  • Deep-sequencing analysis indicates YE1-BE3-nNme2Cas9 converts C residues to T residues at each of the four EGFP target sites.
  • the percentage of editing ranged from 0.24% to 2%.
  • the potential base editing window is from nucleotides 2-8 in the displaced DNA strand, counting the nucleotide at the 5’ (PAM-distal) end as nucleotide #1. See, Figures 3A-D.
  • Figure 3 presents exemplary specific UGI target sites that were respectively integrated into YE1-BE3-nNme2Cas9/D16A mutant fusion proteins and co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line. Converted bases are highlighted in orange color. Background signals were filtered using negative control samples (YE1-BE3- nNme2Cas9 nucleofected K562 cells without sgRNA constructs). N 4 CC PAMs are boxed. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column.
  • Figure 3A shows an exemplary EGFP-Site 1.
  • Figure 3B shows an exemplary EGFP-Site 2.
  • Figure 3C shows an exemplary EGFP-Site 3.
  • Figure 3D shows an exemplary EGFP-Site 4.
  • Electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3- nNme2Cas9 c-fos promoter achieved robust single-base editing of a CxG base pair to a TxA base pair at endogenous target sites in the c-fos promoter (Figure 3E).
  • Figure 3E shows an exemplary deep-sequencing analysis indicating where YE1-BE3-nNme2Cas9 converts C residues to T residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange or yellow color.
  • FIG. 3F shows an exemplary deep-sequencing analysis indicating where ABE7.10-nNme2Cas9 or ABEmax (Koblan et al., 2018)-nNme2Cas9 converts A residues to G residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange color. Background signals were filtered using negative control samples. The percentage of editing is 0.53% by ABE7.10-nNme2Cas9 or 2.33% by ABEmax-nNme2Cas9 (D16A).
  • the present invention contemplates the expression of an ABE7.10- nNme2Cas9 (D16A) fusion protein for base editing.
  • D16A nNme2Cas9
  • Nme2Cas9 base editing may be an effective treatment for tyrosinemia by reversing a G-to-A point mutation in the Fah gene with an
  • ABE7.10-nNme2Cas9 (D16A) fusion protein ABE7.10-nNme2Cas9 (D16A) fusion protein.
  • G-to-A mutation (red) at the last nucleotide of exon 8 in Fah gene, causing exon skipping.
  • FAH deficiency leads to toxin accumulation and severe liver damage.
  • the position of a SpyCas9 PAM (black rectangular box) downstream of the mutation is not optimal for designing the sgRNA since the A mutation is out of the efficient base editing window of ABE7.10, which is 4-7th nt at the 5’ (PAM-distal) end (underlined) (Gaudelli et al., 2017).
  • Nme2Cas9 PAMs red rectangular box
  • D16A ABE7.10-nNme2Cas9
  • Figure 4 presents an exemplary alignment of the wildtype Fah gene with the tyrosinemia Fah mutant gene showing an A-G single base gene editing target site (position 9).
  • the respective SpyCas9 single PAM site and NmeCas9 double PAM sites are indicated for demonstrating the suboptimal targeting window relative to the SpyCas9 PAM site.
  • This figure serves as a potential example of a site where Nme2Cas9 could overcome limitations of existing base editors. It is further believed that the NmeCas9 base editor described herein can perform precise base editing that cannot be achieved with conventional SpyCas9-derived base editors due to a suboptimal base editing window relative to available PAMs nearby.
  • CRISPR Clustered, regularly interspaced, short, palindromic repeats
  • Cas CRISPR-associated proteins
  • CRISPR RNA In Type II CRISPR systems, CRISPR RNA (crRNA) is bound to a trans-activating crRNA (tracrRNA) and loaded onto a Cas9 effector protein that cleaves MGE nucleic acids complementary to the crRNA (Garneau et al., 2010; Deltcheva et al., 2011; Sapranauskas et al., 2011; Gasiunas et al., 2012; Jinek et al., 2012).
  • the crRNA:tracrRNA hybrid can be fused into a single-guide RNA (sgRNA) (Jinek et al., 2012).
  • sgRNA single-guide RNA
  • Cas9 target recognition is usually associated with a 1-5 nucleotide signature downstream of the complementary DNA sequence, called a protospacer adjacent motif (PAM) (Deveau et al., 2008; Mojica et al., 2009).
  • PAM protospacer adjacent motif
  • Streptococcus pyogenes Cas9 (SpyCas9) is the most widely used, in part because it recognizes a short NGG PAM (Jinek et al., 2012) (N represents any nucleotide) that affords a high density of targetable sites. Nevertheless, Spy’s relatively large size (i.e., 1,368 amino acids) makes this Cas9 difficult to package (along with sgRNA and promoters) into a single recombinant adeno- associated virus (rAAV). This has been shown to be a drawback for therapeutic applications given the promise shown by AAV vectors for in vivo gene delivery (Keeler et al., 2017).
  • SpyCas9 and its RNA guides have required extensive characterization and engineering to minimize the tendency to edit near-cognate, off-target sites.
  • subsequent engineering efforts have not overcome these size limitations.
  • NmeCas9, CjeCas9, and GeoCas9 are representatives of type II-C Cas9s (Mir et al., 2018), most of which are ⁇ 1,100 aa. With the exception of GeoCas9, each of these shorter sequence orthologs has been successfully deployed for in vivo editing via all-in-one AAV delivery (in which a single vector expresses both guide and effector) (Ran et al., 2015; Kim et al., 2017; Ibraheim et al., 2018, submitted). Furthermore, NmeCas9 and CjeCas9 have been shown to be naturally resistant to off-target editing (Lee et al., 2016; Kim et al., 2017; Amrani et al., 2018, submitted).
  • SauCas9 mutant (SauCas9 KKH ) has been developed that has reduced PAM constraints (N 3 RRT), though this increase in targeting range often comes at the cost of reduced on-target editing efficacy, and off-target edits are still observed. (Kleinstiver et al., 2015).
  • the present invention contemplates a compact, hyper-accurate Cas9 (Nme2Cas9) from a distinct strain of N. meningitidis.
  • the present invention contemplates a method for single-AAV delivery of Nme2Cas9 and its sgRNA to perform efficient genome editing in vivo and/or ex vivo.
  • this ortholog functions efficiently in mammalian cells and recognizes an N 4 CC PAM that affords a target site density identical to that of wild-type SpyCas9 (e.g., every 8 bp on average, when both DNA strands are considered).
  • PAM Interacting Domains And Anti-CRISPR Proteins PAM recognition by Cas9 orthologs occurs predominantly through protein-DNA interactions between the PAM Interacting Domain (PID) and the nucleotides adjacent to the protospacer (Jiang and Doudna, 2017). PAM mutations often enable phage escape from type II CRISPR immunity (Paez-Espino et al., 2015), placing these systems under selective pressure not only to acquire new CRISPR spacers, but also to evolve new PAM specificities via PID mutations.
  • phages and MGEs express anti-CRISPR (Acr) proteins that inhibit Cas9 (Pawluk et al., 2016; Hynes et al., 2017; Rauch et al., 2017).
  • PID binding is an effective inhibitory mechanism adopted by some Acrs (Dong et al., 2017; Shin et al., 2017; Yang and Patel, 2017), suggesting that PID variation may also be driven by selective pressure to escape Acr inhibition.
  • Cas9 PIDs can evolve such that closely-related orthologs recognize distinct PAMs, as illustrated recently in two species of Geobacillus.
  • the Cas9 encoded by G The Cas9 encoded by G.
  • the present invention contemplates a plurality of N. meninigitidis Cas9 orthologs with divergent PIDs that recognize different PAMs.
  • the present invention contemplates a Cas9 protein with a high sequence identity (>80% along their entire lengths) to that of NmeCas9 strain 8013 (Nme1Cas9) (Zhang et al., 2013). Nme1Cas9 also has a small size and naturally high accuracy as discussed above. (Lee et al., 2016; Amrani et al., 2018).
  • the first clade (group 1) includes orthologs in which the >98% aa sequence identity with Nme1Cas9 extends through the PID.
  • group 2 and group 3 orthologs averaging ⁇ 52% and ⁇ 86% PID sequence identity with Nme1Cas9, respectively.
  • One meningococcal strain was selected from each group: i) De11444 from group 2; and ii) 98002 from group 3 for detailed analysis, which are referred to herein as Nme2Cas9 (1,082 aa) and Nme3Cas9 (1,081 aa), respectively.
  • the CRISPR-cas loci from these two strains have repeat sequences and spacer lengths that are identical to those of strain 8013. See, Figure 6B. This strongly suggested that their mature crRNAs also have 24nt guide sequences and 24 nt repeat sequences (Zhang et al., 2013). Similarly, the tracrRNA sequences of De11444 and 98002 were 100% identical to the 8013 tracrRNA. See, Figure 6B. These observations imply that the same sgRNA sequence scaffold can guide DNA cleavage by all three Cas9s.
  • Nme1Cas9 was replaced with that of either Nme2Cas9 or Nme3Cas9.
  • these protein chimeras were expressed in Escherichia coli, purified, and used for in vitro PAM identification (Karvelis et al., 2015; Ran et al., 2015; Kim et al., 2017). Briefly, a pool of DNA fragments containing a protospacer followed by a 10-nt randomized sequence was cleaved in vitro using recombinant Cas9 and a cognate, in vitro-transcribed sgRNA. See, Figure 5B. Only those DNAs containing a Cas9 PAM sequence were expected to be cleaved. Cleavage products were then sequenced to identify the PAMs. See, Figures 5C-D.
  • ABE7.10-nNme2Cas9 (D16A) is used for single-base editing of AxT base pair to a GxC base pair.
  • BEmax-nNme2Cas9 (D16A) is used for single-base editing of AxT base pair to a GxC base pair.
  • Figure 5 illustrates exemplary three closely related Neisseria meningitidis Cas9 orthologs that have distinct PAMs.
  • Figure 5A shows an exemplary schematic showing mutated residues (orange spheres) between Nme2Cas9 (left) and Nme3Cas9 (right) mapped onto the predicted structure of Nme1Cas9, revealing the cluster of mutations in the PID (black).
  • Figure 5B shows an exemplary experimental workflow of the in vitro PAM discovery assay with a 10-bp randomized PAM region. Following in vitro digestion, adapters were ligated to cleaved products for library construction and sequencing.
  • Figure 5C shows exemplary sequence logos resulting from in vitro PAM discovery reveal the enrichment of a N 4 GATT PAM for Nme1Cas9, consistent with its previously established specificity.
  • Figure 5D shows exemplary sequence logos indicating that Nme1Cas9 with its PID swapped with that of Nme2Cas9 (left) or Nme3Cas9 (right) requires a C at PAM position 5. The remaining nucleotides were not determined with high confidence due to the modest cleavage efficiency of the PID-swapped protein chimeras (see Figure 6C).
  • Figure 5E shows an exemplary sequence logo showing that full-length Nme2Cas9 recognizes an N 4 CC PAM, based on efficient substrate cleavage of a target pool with a fixed C at PAM position 5, and with PAM nts 1-4 and 6-8 randomized.
  • Figure 6 presents a characterization of Neisseria meningitidis Cas9 orthologs with rapidly-evolving PIDs, as related to Figure 5.
  • Figure 6A shows an exemplary unrooted phylogenetic tree of NmeCas9 orthologs that are >80% identical to Nme1Cas9. Three distinct branches emerged, with the majority of mutations clustered in the PID. Groups 1 (blue), 2 (orange), and 3 (green) have PIDs with >98%, approximately 52%, and approximately86% identity to Nme1Cas9, respectively. Three representative Cas9 orthologs (one from each group) (Nme1Cas9, Nme2Cas9 and Nme3Cas9) are indicated.
  • Figure 6B shows an exemplary schematic showing the CRISPR-cas loci of the strains encoding the three Cas9 orthologs (Nme1Cas9, Nme2Cas9, and Nme3Cas9) from (A). Percent identities of each CRISPR-Cas component with N. meningitidis 8013 (encoding Nme1Cas9) are shown. Blue and red arrows denote pre-crRNA and tracrRNA transcription initiation sites, respectively.
  • Figure 6C shows an exemplary normalized read counts (% of total reads) from cleaved DNAs from the in vitro assays for intact Nme1Cas9 (grey), for chimeras with Nme1Cas9’s PID swapped with those of
  • Nme2Cas9 and Nme3Cas9 are plotted.
  • the reduced normalized read counts indicate lower cleavage efficiencies in the chimeras.
  • Figure 6D shows an exemplary sequence logos from the in vitro PAM discovery assay on an
  • Nme2Cas9 To test the efficacy of Nme2Cas9 in human genome editing, a full-length (e.g., not PID- swapped) human-codon-optimized Nme2Cas9 construct was cloned into a mammalian expression plasmid with appended nuclear localization signals (NLSs) and linkers validated previously for Nme1Cas9 (Amrani et al., 2018). For initial tests, a modified, fluorescence-based Traffic Light Reporter (TLR2.0) was used (Certo et al., 2011). Briefly, a disrupted GFP is followed by an out-of-frame T2A peptide and mCherry cassette.
  • NLSs nuclear localization signals
  • TLR2.0 Fluorescence-based Traffic Light Reporter
  • Homology-directed repair (HDR) outcomes can also be scored simultaneously by including a DNA donor that restores the functional GFP sequence, yielding a green fluorescence (Certo et al., 2011). Because some indels do not introduce a +1 frameshift, the fluorescence readout generally provides an underestimate of the true editing efficiency. Nonetheless, the speed, simplicity, and low cost of the assay makes it useful as an initial, semi-quantitative measure of genome editing in HEK293T cells carrying a single TLR2.0 locus incorporated via lentivector.
  • Nme2Cas9 plasmid was transiently co-transfected with one of fifteen sgRNA plasmids carrying spacers that target TLR2.0 sites with N 4 CC PAMs. No HDR donor was included, so only NHEJ-based editing (mCherry) was scored. Most sgRNAs were in a G23 format (i.e. a 5’-terminal G to facilitate transcription, followed by a 23nt guide sequence), as used routinely for Nme1Cas9 (Lee et al., 2016; Pawluk et al., 2016; Amrani et al., 2018;
  • Nme2Cas9 all 15 targets with N 4 CC PAMs were functional, though to various extents ranging from 4% to 20% mCherry. These fifteen sites include examples with each of the four possible nucleotides in the 7 th PAM position (e.g., after the CC dinucleotide), indicating that a slight preference for an A residue was observed in vitro (Figure 5E) does not reflect a PAM requirement for editing applications in human cells.
  • the N 4 GATT PAM control yielded mCherry signal similar to no-sgRNA control. See, Figure 7B.
  • N 4 DC A, T, G
  • N 4 CD PAM sites were tested in TLR2.0 reporter cells. See, Figures 8A and 8B. No detectable editing was found at any of these sites, providing an initial indication that both C residues of the N 4 CC PAM consensus are required for efficient Nme2Cas9 activity.
  • the length of the spacer in the crRNA differs among Cas9 orthologs and can affect on- vs. off-target activity (Cho et al., 2014; Fu et al., 2014).
  • SpyCas9 optimal spacer length is 20 nts, with truncations down to 17 nts tolerated (Fu et al., 2014).
  • Nme1Cas9 usually has 24-nt spacers (Hou et al., 2013; Zhang et al., 2013), and tolerates truncations down to 18-20 nts (Lee et al., 2016; Amrani et al., 2018).
  • Figure 7 presents exemplary data showing that Nme2Cas9 uses a 22-24 nt spacer to edit sites adjacent to an N 4 CC PAM. All experiments were done in triplicate, and error bars represent the standard error of the mean (s.e.m.).
  • Figure 7A shows an exemplary schematic diagram depicting transient transfection and editing of HEK293T TLR2.0 cells, with mCherry+ cells detected by flow cytometry 72 hours after transfection.
  • Figure 7B shows an exemplary
  • Nme2Cas9 editing of the TLR2.0 reporter Sites with N 4 CC PAMs were targeted with varying efficiencies, while no Nme2Cas9 targeting was observed at an N 4 GATT PAM or in the absence of sgRNA.
  • Figure 7C shows an exemplary effect of spacer length on the efficiency of Nme2Cas9 editing.
  • FIG. 7D shows an exemplary An Nme2Cas9 dual nickase can be used in tandem to generate NHEJ- and HDR-based edits in TLR2.0.
  • Cleavage sites 32 bp and 64 bp apart were targeted using either nickase.
  • the HNH nickase (Nme2Cas9 D16A ) yielded efficient editing, particularly with the cleavage sites that were separated by 32 bp, whereas the RuvC nickase (Nme2Cas9 H588A ) was not effective. Wildtype Nme2Cas9 was used as a control.
  • Cas9 enzymes use their HNH and RuvC domains to cleave the guide-complementary and non-complementary strand of the target DNA, respectively.
  • SpyCas9 nickases in which either the HNH or RuvC domain is mutationally inactivated, have been used to induce homology-directed repair (HDR) and to improve genome editing specificity via DSB induction by dual nickases (Mali et al., 2013a; Ran et al., 2013).
  • Nme2Cas9 D16A HNH nickase
  • Nme2Cas9 H588A RuvC nickase
  • Target sites within TLR2.0 were used to test the functionality of each nickase using guides targeting cleavage sites spaced 32 bp and 64 bp apart. See, Figure 7D. Wildtype Nme2Cas9 targeting a single site showed efficient editing, with both NHEJ and HDR as outcomes of repair. For nickases, cleavage sites 32 bp and 64 bp apart showed editing using the Nme2Cas9 D16A (HNH nickase), but neither target pair worked with Nme2Cas9 H588A . These results suggest that Nme2Cas9 HNH nickase can be used for efficient genome editing, as long as the sites are in close proximity.
  • Figure 8 presents exemplary data showing PAM, spacer, and seed requirements for Nme2Cas9 targeting in mammalian cells, as related to Figure 7. All experiments were done in triplicate and error bars represent s.e.m.
  • Figure 8A shows an exemplary Nme2Cas9 targeting at N 4 CD sites in TLR2.0, with editing estimated based on mCherry+ cells. Four sites for each non- C nucleotide at the tested position (N 4 CA, N 4 CT and N 4 CG) were examined, and an N 4 CC site was used as a positive control.
  • Figure 8B shows an exemplary Nme2Cas9 targeting at N 4 DC sites in TLR2.0 [similar to (A)].
  • Figure 8C shows exemplary guide truncations on a TLR2.0 site (distinct from that in Figure 2C) with a N 4 CCA PAM, revealing similar length requirements as those observed at the other site.
  • Figure 8D shows exemplary Nme2Cas9 targeting efficiency is differentially sensitive to single-nucleotide mismatches in the seed region of the sgRNA. Data show the effects of walking single-nucleotide sgRNA mismatches along the 23-nt spacer in a TLR2.0 target site.
  • Nme2Cas9 s ability to function in different mammalian cell lines was tested using various delivery methods. As an initial test, forty (40) different sites (29 with a N 4 CC PAM, and 11 sites were tested with a N 4 CD PAM). Several loci were selected (AAVS1, VEGFA, etc.), and target sites with N 4 CC PAMs were randomly chosen for editing with Nme2Cas9. Editing (%) was determined by transiently transfecting 150 ng of Nme2Cas9 along with 150 ng of sgRNA plasmids followed by TIDE analysis 72 hours post-transfection. A subset of sites exhibiting a range of editing efficiencies in this initial screen was selected for repeat analyses in triplicate. See, Figure 9A; and Table 1.
  • Figure 9 presents exemplary data showing Nme2Cas9 genome editing at endogenous loci in mammalian cells via multiple delivery methods. All results represent 3 independent biological replicates, and error bars represent s.e.m.
  • Figure 9A shows an exemplary Nme2Cas9 genome editing of endogenous human sites in HEK293T cells following transient transfection of Nme2Cas9- and sgRNA-expressing plasmids.40 sites were screened initially (Table 1); the 14 sites shown (selected to include representatives of varying editing efficiencies, as measured by TIDE) were then re-analyzed in triplicate. An Nme1Cas9 target site (with an N 4 GATT PAM) was used as a negative control.
  • Figure 9B shows exemplary data charts: Left panel: Transient transfection of a single plasmid expressing both Nme2Cas9 and sgRNA (targeting the Pcsk9 and Rosa26 loci) enables editing in Hepa1-6 mouse cells, as detected by TIDE. Right panel:
  • FIG. 9C shows exemplary Nme2Cas9 can be electroporated as an RNP complex to induce genome editing.40 picomoles Cas9 along with 50 picomoles of in vitro-transcribed sgRNAs targeting three different loci were electroporated into HEK293T cells. Indels were measured after 72h using TIDE. Table 1. Exemplary Endogenous human genome editing sites targeted by Nme2Cas9.
  • HEK293T cells were used to support transient transfections and at 72-hours post transfection the, cells were harvested, followed by genomic DNA extraction and selective amplification of the targeted locus. TIDE analysis was used to measure indel efficiency at each locus (Brinkman et al., 2014). Nme2Cas9 editing was detectable at most of these sites, even though efficiencies varied depending on the target sequence. Table 1. Interestingly, Nme2Cas9 induced indels at several genomic sites with N 4 CD PAMs, albeit less consistently and at lower levels. Table 1. Fourteen (14) sites with N 4 CC PAMs were analyzed in triplicate, and consistent editing was observed. See, Figure 9A. In addition, editing efficiency could be improved significantly by increasing the quantity of the Nme2Cas9 plasmid delivered, and this high efficiency could be extended to precise segmental deletion with two guides. See, Figures 10A and 10B.
  • Nme2Cas9 The ability of Nme2Cas9 to function was tested in mouse Hepa1-6 cells (hepatoma- derived). For Hepa1-6 cells, a single plasmid encoding both Nme2Cas9 and an sgRNA (targeting either Rosa26 or Pcsk9) was transiently transfected and indels were measured after 72 hrs.
  • Nme2Cas9 s functionality was also tested when stably expressed in human leukemia K562 cells.
  • a lentiviral construct was created expressing Nme2Cas9 and transduced cells to stably express Nme2Cas9 under the control of the SFFV promoter.
  • This stable cell line did not show any visible differences with respect to growth and morphology in comparison to untransduced cells, suggesting that Nme2Cas9 is not toxic when stably expressed.
  • These cells were transiently electroporated with plasmids expressing sgRNAs and analyzed by TIDE after 72 hours to measure indel efficiencies. Efficient (>50%) editing was observed at all three sites tested, validating Nme2Cas9’s ability to function upon lentiviral delivery in K562 cells. See, Figure 9B.
  • Ribonucleoprotein (RNP) delivery of Cas9 and its sgRNA is also useful for some genome editing applications, and the greater transience of Cas9’s presence can minimize off-target editing (Kim et al., 2014; Zuris et al., 2015). Moreover, some cell types (e.g. certain immune cells) are recalcitrant to DNA transfection-based editing (Schumann et al., 2015).
  • Nme2Cas9 is functional by RNP delivery, a 6xHis-tagged Nme2Cas9 (fused to three NLSs) was cloned into a bacterial expression construct and the recombinant protein was purified.
  • Nme2Cas9 can be delivered effectively via plasmid or lentivirus, or as an RNP complex, in multiple cell types.
  • Nme2Cas9 Considering the high sequence identity between Nme1Cas9 and Nme2Cas9, at least some of these Acr families should inhibit Nme2Cas9. To test this, all five families of recombinant Acrs were expressed, purified and tested for Nme2Cas9’s ability to cleave a target in vitro in the presence of a member of each family (10:1 Acr:Cas9 molar ratio). An inhibitor was used for the type I-E CRISPR system in E. coli (AcrE2) as a negative control, while Nme1Cas9was used as a positive control. (Pawluk et al., 2014); (Pawluk et al., 2016). As expected, all 5 families inhibited Nme1Cas9, while AcrE2 failed to do so. See, Figure 11A, top. AcrIIC1 Nme , AcrIIC2 Nme ,
  • Figure 10 presents exemplary data showing dose dependence and segmental deletions by Nme2Cas9, as related to Figure 9.
  • Figure 10A shows exemplary increasing the dose of electroporated Nme2Cas9 plasmid (500 ng, vs.200 ng in Figure 3A) improves editing efficiency at two sites (TS16 and TS6). Data provided in yellow are re-used from Figure 9A.
  • Figure 10B shows exemplary Nme2Cas9 can be used to create precise segmental deletions. Two TLR2.0 targets with cleavage sites 32 bp apart were targeted simultaneously with Nme2Cas9. The majority of lesions created were deletions of exactly 32 bp (blue).
  • Figure 11 presents exemplary data showing that Nme2Cas9 is subject to inhibition by a subset of type II-C anti-CRISPR families in vitro and in cells. All experiments were done in triplicate and error bars represent s.e.m.
  • Figure 11A shows exemplary In vitro cleavage assay of Nme1Cas9 and Nme2Cas9 in the presence of five previously characterized anti-CRISPR proteins (10:1 ratio of Acr:Cas9). Top: Nme1Cas9 efficiently cleaves a fragment containing a protospacer with an N 4 GATT PAM in the absence of an Acr or in the presence of a negative control Acr (AcrE2). All five previously characterized type II-C Acr families inhibited
  • Nme1Cas9 As expected. Bottom: Nme2Cas9 inhibition mirrors that of Nme1Cas9, except for the lack of inhibition by AcrIIC5 Smu .
  • Figure 11B shows exemplary genome editing in the presence of the five previously described anti-CRISPR families. Plasmids expressing Nme2Cas9 (200 ng), sgRNA (100 ng) and each respective Acr (200 ng) were co-transfected into HEK293T cells, and genome editing was measured using Tracking of Indels by Decompostion (TIDE) 72 hr post transfection.
  • TIDE Decompostion
  • FIG. 11C shows exemplary Acr inhibition of Nme2Cas9 is dose-dependent with distinct apparent potencies. Nme2Cas9 is fully inhibited by AcrIIC1 Nme and AcrIIC4 Hpa at 2:1 and 1:1 mass ratios of cotransfected Acr and Nme2Cas9 plasmids, respectively.
  • Nme1Cas9/Nme2Cas9 chimera with the PID of Nme2Cas9 was tested. See, Figure 5D and Figure 6D. Due to the reduced activity of this hybrid, a ⁇ 30x higher concentration of Cas9 was used to achieve a similar cleavage efficiency while maintaining the 10:1 Cas9:Acr molar ratio. No inhibition was observed by AcrIIC5 Smu on this protein chimera. See, Figure 12. This data provides further evidence that AcrIIC5 Smu likely interacts with the PID of Nme1Cas9. Regardless of the mechanistic basis for the differential inhibition by AcrIIC5 Smu , these results indicate that Nme2Cas9 is subject to inhibition by the other four type II-C Acr families.
  • Figure 12 presents exemplary data showing that a Nme2Cas9 PID swap renders
  • Nme1Cas9 insensitive to AcrIIC5 Smu inhibition as related to Figure 11.
  • Nme1Cas9-Nme2Cas9PID chimera in the presence of previously characterized Acr proteins (10 uM Cas9-sgRNA + 100 uM Acr).
  • Nme2Cas9/sgRNA plasmid transfections 150 ng of each plasmid targeting TS16 were performed in HEK293T cells in the presence or absence of Acr expression plasmids, as it has been reported that most Acrs inhibited Nme1Cas9 at those plasmid ratios (Pawluk et al., 2016).
  • Nme1Cas9 demonstrates remarkable editing fidelity in cells and mouse models (Lee et al., 2016; Amrani et al., 2018; Ibraheim et al., 2018). Furthermore, the similarity of Nme2Cas9 to Nme1Cas9 over most of its length suggests that it may likewise be hyper-accurate. However, the higher number of sites sampled in the genome as a result of the dinucleotide PAM could create more opportunities for Nme2Cas9 off-targeting in comparison with Nme1Cas9 and its less frequently encountered 4-nucleotide PAM.
  • GUIDE-seq gene-wide, unbiased identification of double-stranded breaks enabled by sequencing was used to identify potential off-target sites empirically and in an unbiased fashion (Tsai et al., 2014). Even the best off-target prediction algorithms are prone to false negatives necessitating empirical target site profiling methods (Bolukbasi et al., 2015b; Tsai and Joung, 2016; Tycko et al., 2016).
  • GUIDE-seq relies on the incorporation of double-stranded oligodeoxynucleotides (dsODNs) into DNA double-stranded break sites throughout the genome. These insertion sites are then detected by amplification and high-throughput sequencing.
  • dsODNs double-stranded oligodeoxynucleotides
  • SpyCas9 is a well-characterized Cas9 ortholog it is useful for multiplexed applications with other Cas9s, and as a benchmark for their editing properties (Jiang and Doudna, 2017; Komor et al., 2017).
  • SpyCas9 and Nme2Cas9 were cloned into identical plasmid backbones, with the same UTRs, linkers, NLSs, and promoters, for parallel transient transfections (along with similarly matched sgRNA-expressing plasmids) into HEK293T cells.
  • the RNA guides for SpyCas9 and Nme2Cas9 are orthogonal, i.e.
  • Nme2Cas9 sgRNAs do not direct editing by SpyCas9, and vice versa. See, Figure 13A. This was in contrast to earlier reported results with Nme1Cas9 (Esvelt et al., 2013; Fonfara et al., 2014).
  • DS2, DS4 and DS6 were selected to sample off-target cleavage with Nme2Cas9 guides that direct on-target editing as efficiently, less efficiently, or more efficiently than the corresponding SpyCas9 guides, respectively.
  • TS6 was added as it has been observed to be an efficiently edited Nme2Cas9 target sites, having an approximate 30-50% indel efficiency depending on the cell type. See, Figures 9A and 10A. Similar data is seen with the mouse Pcsk9 and Rosa26 Nme2Cas9 sites. See, Figure 9B.
  • GUIDE-seq libraries were prepared as described previously (Amrani et al., 2018).
  • a GUIDE-seq analysis revealed efficient on-target editing for both Cas9 orthologs, with relative efficiencies (as reflected by GUIDE-seq read counts) that are similar to those observed by TIDE.
  • Figure 13B and Table 2. (Tsai et al., 2014; Zhu et al., 2017).
  • Figure 13 presents exemplary data showing orthogonality and relative accuracy of Nme2Cas9 and SpyCas9 at dual target sites, as related to Figure 12.
  • Figure 13A shows exemplary Nme2Cas9 and SpyCas9 guides are orthogonal. TIDE results show the frequencies of indels created by both nucleases targeting DS2 with either their cognate sgRNAs or with the sgRNAs of the other ortholog.
  • Figure 13B shows exemplary Nme2Cas9 and SpyCas9 exhibiting comparable on-target editing efficiencies as assessed by GUIDE-seq. Bars indicate on-target read counts from GUIDE-Seq at the three dual sites targeted by each ortholog.
  • Orange bars represent Nme2Cas9 and black bars represent SpyCas9.
  • Figure 13C shows an exemplary SpyCas9’s on- target vs. off-target read counts for each site. Orange bars represent the on-target reads while black bars represent off-targets.
  • Figure 13D shows exemplary Nme2Cas9’s on-target vs. off- target reads for each site.
  • Figure 13E bar graphs showing exemplary indel efficiencies (measured by TIDE) at potential off-target sites predicted by CRISPRSeek. On- and off-target site sequences are shown on the left, with the PAM region underlined and sgRNA mismatches and non-consensus PAM nucleotides given in red. Table 2: GUIDE-seq Data For off-target identification, the analysis revealed that the DS2, DS4, and DS6
  • SpyCas9 sgRNAs appeared to direct editing at 93, 10, and 118 candidate off-target sites, respectively, in the normal range of off-targets when plasmid-based SpyCas9 editing is analyzed by GUIDE-seq (Fu et al., 2014; Tsai et al., 2014).
  • the DS2, DS4, and DS6 Nme2Cas9 sgRNAs appeared to direct editing at 1, 0, and 1 off-target sites, respectively.
  • Figure 14C and Table 2 When compared to the GUIDE-seq read counts for the SpyCas9 off-targets, those of Nme2Cas9 were very low, further suggesting that Nme2Cas9 is highly specific.
  • Figure 13C cf.
  • Figure 13D Nme2Cas9 GUIDE-seq analyses with the TS6, Pcsk9, and Rosa26 yielded similar results (0, 0, and 1 off-target sites, respectively, with a modest read count for the Rosa26-OT1 off-target site).
  • Figure 13C, Figure 14D, and Table 2 Nme2Cas9 GUIDE-seq analyses with the TS6, Pcsk9, and Rosa26 yielded similar results (0, 0, and 1 off-target sites, respectively, with a modest read count for the Rosa26-OT1 off-target site).
  • Figure 14 presents exemplary data showing that Nme2Cas9 exhibits little or no detectable off-targeting in mammalian cells.
  • Figure 14A shows an exemplary schematic depicting dual sites (DSs) targetable by both SpyCas9 and Nme2Cas9 by virtue of their non-overlapping PAMs. The Nme2Cas9 PAM (orange) and SpyCas9 PAM (blue) are highlighted. A 24nt Nme2Cas9 guide sequence is indicated in yellow; the corresponding guide sequence for SpyCas9 would be 4nt shorter at the 5’ end.
  • Figure 14B shows an exemplary Nme2Cas9 and SpyCas9 that both induce indels at DSs.
  • Nme2Cas9 (DS1 and DS4).
  • DS2, DS4 and DS6 were selected for GUIDE-Seq analysis as Nme2Cas9 was equally efficient, less efficient and more efficient than SpyCas9, respectively, at these sites.
  • Figure 14C shows exemplary Nme2Cas9 genome editing that is highly accurate in human cells. Numbers of off-target sites detected by GUIDE-Seq for each nuclease at individual target sites are shown. In addition to dual sites, we analyzed TS6 (because of its high on-target editing efficiency) and Pcsk9 and Rosa26 sites in mouse Hepa1-6 cells (to measure accuracy in another cell type).
  • Figure 14D shows an exemplary targeted deep sequencing to detect indels in edited cells confirms the high Nme2Cas9 accuracy indicated by GUIDE-seq.
  • Figure 14E shows an exemplary sequence for the validated off-target site of the Rosa26 guide, showing the PAM region (underlined), the consensus CC PAM dinucleotide (bold), and three mismatches in the PAM-distal portion of the spacer (red).
  • Nme2Cas9 offer major advantages for in vivo genome editing using Associated Adenovirus (AAV) delivery.
  • AAV Associated Adenovirus
  • Nme2Cas9 was cloned with its sgRNA and their promoters (U1a and U6, respectively) into an AAV vector backbone. See, Figure 15A.
  • An all-in-one AAV was prepared with an sgRN- .Nme2Cas9 packaged into a hepatotropic AAV8 capsid to target two genes in the mouse liver: i) Rosa26 (a commonly used safe harbor locus for transgene insertion) (Friedrich and Soriano, 1991) as a negative control; and ii) Pcsk9, a major regulator of circulating cholesterol homeostasis (Rashid et al., 2005), as a phenotypic target.
  • GCs AAV8.sgRNA.Nme2Cas9 genome copies
  • Figure 15 presents exemplary data showing Nme2Cas9 genome editing in vivo via all-in- one AAV delivery.
  • Figure 15A shows exemplary workflow for delivery of
  • AAV8.sgRNA.Nme2Cas9 to lower cholesterol levels in mice by targeting Pcsk9.
  • Top schematic of the all-in-one AAV vector expressing Nme2Cas9 and the sgRNA (individual genome elements not to scale).
  • BGH bovine growth hormone poly(A) site
  • HA epitope tag
  • NLS nuclear localization sequence
  • h human-codon-optimized.
  • FIG. 15B shows an exemplary TIDE analysis to measure indels in DNA extracted from livers of mice injected with AAV8.Nme2Cas9+sgRNA targeting Pcsk9 and Rosa26 (control) loci. Indel efficiency at the lone off-target site identified by GUIDE-seq for these two sgRNAs (Rosa26
  • Figure 15C shows an exemplary reduced serum cholesterol levels in mice injected with the Pcsk9-targeting guide compared to the Rosa26- targeting controls.
  • Figure 16 presents exemplary data showing PCSK9 knockdown and liver histology following Nme2Cas9 AAV delivery and editing, related to Figure 15.
  • Figure 16A shows exemplary Western blotting using anti-PCSK9 antibody reveals strongly reduced levels of PCSK9 in the livers of mice treated with sgPcsk9, compared to mice treated with sgRosa26.2ng of recombinant PCSK9 was used as a mobility standard (left-most lane), and a cross-reacting band in the liver samples is indicated by an asterisk. GAPDH was used as loading control (bottom panel).
  • Figure 16B shows exemplary H&E staining from livers of mice injected with AAV8.Nme2Cas9+sgRosa26 (left) or
  • AAV vectors have recently been used for the generation of genome-edited mice, without the need for microinjection or electroporation, simply by soaking the zygotes in culture medium containing AAV vector(s), followed by reimplantation into pseudopregnant females (Yoon et al., 2018). Editing was obtained previously with a dual-AAV system in which SpyCas9 and its sgRNA were delivered in separate vectors (Yoon et al., 2018). To test whether Nme2Cas9 could perform accurate and efficient editing in mouse zygotes with an all - in-one AAV delivery system, we targeted Tyrosinase (Tyr). A bi-allelic inactivation of Tyr disrupts melanin production resulting in an albino phenotype (Yokoyama et al., 1990).
  • Figure 17A shows an exemplary two sites in Tyr, each with N 4 CC PAMs, were tested for editing in Hepa1-6 cells. The sgTyr2 guide exhibited higher editing efficiency and was selected for further testing.
  • Figure 17B shows an exemplary seven mice that survived post-natal development, and each exhibited coat color phenotypes as well as on-target editing, as assayed by TIDE.
  • Figure 17C shows an exemplary Indel spectra from tail DNA of each mouse from (B), as well as an unedited C57BL/6NJ mouse, as indicated by TIDE analysis. Efficiencies of insertions (positive) and deletions (negative) of various sizes are indicated.
  • Figure 18 presents exemplary data showing Nme2Cas9 genome editing ex vivo via all-in- one AAV delivery.
  • Figure 18A shows an exemplary workflow for single-AAV Nme2Cas9 editing ex vivo to generate albino C57BL/6NJ mice by targeting the Tyr gene.
  • Zygotes are cultured in KSOM containing AAV6.Nme2Cas9:sgTyr for 5-6 hours, rinsed in M2, and cultured for a day before being transferred to the oviduct of pseudo-pregnant recipients.
  • Figure 18B shows exemplary albino (left) and chinchilla or variegated (middle) mice generated by 3x10 9 GCs, and chinchilla or variegated mice (right) generated by 3x10 8 GCs of zygotes with
  • FIG. 18C shows an exemplary summary of Nme2Cas9.sgTyr single- AAV ex vivo Tyr editing experiments at two AAV doses.
  • mice 9-2 The data is inconclusive as to whether there was no mosaicism in mouse 9-2, or that additional alleles were absent from mouse 9-1, because only tail samples were sequenced and other tissues could have distinct lesions.
  • Analysis of tail DNA from chinchilla mice revealed the presence of in-frame mutations that are potentially the cause of the chinchilla coat color.
  • the limited mutational complexity suggests that editing occurred early during embryonic development in these mice.
  • Figure 19 shows an exemplary mCherry reporter assay for nSpCas9-ABEmax and optimized nNme2Cas9-ABEmax activities.
  • Figure 19A shows exemplary sequence information of ABE-mCherry reporter. There is a TAG stop codon in the mCherry coding region. In the reporter-integrated stable cell line, there is no mCherry signal due to this stop codon. The mCherry signal will be activated if the nSpCas9-ABEmax or optimized nNme2Cas9-ABEmax can convert TAG to CAG, which encodes a glutamine residue.
  • Figure 19B shows an exemplary mCherry signal is activated due to SpCas9-ABE or Nme2Cas9-ABE activity.
  • Figure 20 shows an exemplary GFP reporter assay for nSpCas9-CBE4 (Addgene #100802) and nNme2Cas9-CBE4 (same plasmid backbone as Addgene #100802) activities.
  • Figure 20A shows exemplary sequence information of the CBE-GFP reporter. There is a mutation that converts GYG to GHG in the fluorophore core region of the GFP reporter line. There is no GFP signal due to this mutation. The GFP signal will be activated if the nSpCas9- CBE4 or nNme2Cas9-CBE4 can convert CAC (encoding histidine) to TAC/TAT (encoding tyrosine).
  • Figure 20B shows an exemplary GFP signal is activated due to nSpCas9-CBE4 or nNme2Cas9-CBE4 activity.
  • Upper panel negative control (no editing); middle panel: GFP activation by nSpCas9-CBE4; bottom panel: GFP activation by nNme2Cas9-CBE4).
  • Figure 20C shows an exemplary FACS quantitation of base editing events in GFP reporter cells transfected with nSpCas9-CBE4 or nNme2Cas9-CBE4.
  • N 6; error bars represent S.D. Results are from biological replicates performed in technical duplicates.
  • Figure 21 shows exemplary cytosine editing by nNme2Cas9-CBE4.
  • Upper panel shows the KANK3 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples.
  • Bottom panel shows the quantification of the substitution efficiency of each type of base in the nNmeCas9-CBE4 editing window of the KANK3 target sequences.
  • Sequence tables show nucleotide frequencies at each position.
  • Figure 22 shows exemplary cytosine and adenine editing by nNme2Cas9-CBE4 and
  • nNme2Cas9-ABEmax nNme2Cas9-ABEmax, respectively.
  • Upper panel shows the PLXNB2 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples.
  • Middle panel shows the quantification of the substitution rate of each type of base in the nNmeCas9-ABEmax editing windows of the PLXNB2 target sequence.
  • Sequence tables show nucleotide frequencies at each position. Frequencies of expected A-to-G conversion are highlighted in red.
  • Bottom panel shows the quantification of the substitution efficiency of each type of base in the nNmeCas9-CBE4 editing windows of the PLXNB2 target sequence. Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red.
  • Non-PID aa differences (teal- underlined); PID aa differences (yellow - underlined bold); active site residues (red - bold).
  • NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFD ED Nme1Cas9 (661-720)
  • Non-PID aa differences (teal- underlined); PID aa differences (yellow - underlined bold); active site residues (red - bold).
  • Nme1Cas91 MAAFKPNSINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAE 50
  • Nme3Cas91 MAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAE 50 Nme1Cas951
  • Nme3Cas9351 KAYHTISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK 400
  • Nme1Cas9401 DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG 450
  • Nme3Cas9401 DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG 450
  • Nme1Cas9451 DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR 500
  • Nme3Cas9451 DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR 500 Nme1Cas9501 IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS 550
  • Nme3Cas9501 IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS 550 Nme1Cas9551
  • NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ 650 Nme3Cas9601
  • NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ 650 Nme1Cas9651
  • Nme3Cas91050 GVKTALSFQKYQIDEMGKEIRPCRLKKRPPVR 1081 Plasmid-Expressed Nme2Cas9
  • SV40 NLS yellow- BOLD
  • 3X-HA-Tag green-(underlined/bold
  • cMyc-like NLS teal- plain
  • Linker magenta - bold italics
  • Nme2Cas9 italics
  • SV40 NLS (yellow- BOLD); 3X-HA-Tag (green-(underlined/bold); Nucleoplasmin-like NLS (red-underline); c-myc NLS (teal- plain); Linker (magenta - bold italics) and Nme2Cas9 (italics).
  • SV40 NLS yellow- BOLD
  • Nucleoplasmin-like NLS red-underline
  • Linker magenta - bold italics
  • Nme2Cas9 italics
  • Nme2Cas9 as a compact and highly accurate Cas9 with a less restrictive dinucleotide PAM for genome editing by AAV delivery in vivo.
  • the development of Nme2Cas9 greatly expands the genomic scope of in vivo editing, especially via viral vector delivery.
  • the Nme2Cas9 all-in-one AAV delivery platform established in this study can in principle be used to target as wide a range of sites as SpyCas9 (due to the identical densities of optimal N 4 CC and NGG PAMs), but without the need to deliver two separate vectors to the same target cells.
  • Nme2Cas9 a catalytically dead version of Nme2Cas9
  • dNme2Cas9 a catalytically dead version of Nme2Cas9
  • CRISPRi CRISPRa
  • base editing and related approaches
  • Nme2Cas9 hyper-accuracy enables precise editing of target genes, potentially ameliorating safety issues resulting from off-target activities.
  • Nme2Cas9 compared to that of Nme1Cas9
  • PAMs PAMs
  • Type II-C Cas9 orthologs are generally slower nucleases in vitro than SpyCas9 (Ma et al., 2015; Mir et al., 2018); interestingly, enzymological principles indicate that a reduced apparent k cat (within limits) can improve on- vs. off-target specificity for RNA-guided nucleases (Bisaria et al., 2017).
  • Nme2Cas9 and Nme3Cas9 hinged on unexplored Cas9s that are highly related (outside of the PID) to an ortholog that was previously validated for human genome editing (Esvelt et al., 2013; Hou et al., 2013; Lee et al., 2016; Amrani et al., 2018).
  • the relatedness of Nme2Cas9 and Nme3Cas9 to Nme1Cas9 brought an added benefit, namely that they use the exact same sgRNA scaffold, circumventing the need to identify and validate functional tracrRNA sequences for each.
  • Cas9 inhibitors such as AcrIIC1 that bind more conserved regions of Cas9 likely present fewer routes toward mutational escape and therefore exhibit a broader inhibitory spectrum (Harrington et al., 2017a).
  • Nme2Cas9 e.g. AcrIIC1-4
  • the approach used in this study i.e. searching for rapidly-evolving domains within Cas9 can be implemented elsewhere, especially with bacterial species that are well-sampled at the level of genome sequence.
  • This approach could also be applied to other CRISPR-Cas effector proteins such as Cas12 and Cas13 that have also been developed for genome or transcriptome engineering and other applications.
  • This strategy could be especially compelling with Cas proteins that are closely related to orthologs with proven efficacy in heterologous contexts (e.g. in eukaryotic cells), as was the case for Nme1Cas9.
  • the application of this approach to meningococcal Cas9 orthologs yielded a new genome editing platform, Nme2Cas9, with a unique combination of characteristics (compact size, dinucleotide PAM, hyper-accuracy, single- AAV deliverability, and Acr susceptibility) that promise to accelerate the development of genome editing tools for both general and therapeutic applications.
  • Table 3 The following presents exemplary sequences for plasmids and oligos as disclosed herein.
  • the Neon electroporation system was used exactly as described (Amrani et al., 2018). Briefly, 40 picomoles of 3xNLS-Nme2Cas9 along with 50 picomoles of T7-transcribed sgRNA was assembled in buffer R and electroporated using 10 mL Neon tips. After electroporation, cells were plated in pre-warmed 24-well plates containing the appropriate culture media without antibiotics. Electroporation parameters (voltage, width, number of pulses) were 1150 V, 20 ms, 2 pulses for HEK293T cells; 1000 V, 50 ms, 1 pulse for K562 cells.
  • AAV8.Nme2Cas9+sgRNA delivery and liver tissue processing For the AAV8 vector injections, 8-week-old female C57BL/6NJ mice were injected with 4 x10 11 genome copies per mouse via tail vein, with the sgRNA targeting a validated site in either Pcsk9 or Rosa26. Mice were sacrificed 28 days after vector administration and liver tissues were collected for analysis. Liver tissues were fixed in 4% formalin overnight, embedded in paraffin, sectioned and stained with hematoxylin and eosin (H&E). Blood was drawn from the facial vein at 0, 14 and 28 days post injection, and serum was isolated using a serum separator (BD, Cat. No.365967) and stored at -80°C until assay. Serum cholesterol level was measured using the InfinityTM colorimetric endpoint assay (Thermo-Scientific) following the
  • KSOM Potassium-Supplemented Simplex Optimized Medium, Millipore, Cat. No. MR-106-D
  • AAV6.Nme2Cas9.sgTyr vector for 5-6 h (4 zygotes in each drop). After incubation, zygotes were rinsed in M2 and transferred to fresh KSOM for overnight culture. The next day, the embryos that advanced to 2-cell stage were transferred into the oviduct of pseudopregnant recipients and allowed to develop to term.
  • Nme1Cas9 peptide sequence was used as a query in BLAST searches to find all Cas9 orthologs in Neisseria meningitidis species. Orthologs with >80% identity to Nme1Cas9 were selected for the remainder of this study. The PIDs were then aligned with that of Nme1Cas9 (residues 820-1082) using ClustalW2 and those with clusters of mutations in the PID were selected for further analysis. An unrooted phylogenetic tree of NmeCas9 orthologs was constructed using FigTree (http://tree.bio.ed.ac.uk/software/figtree/).
  • Rosetta (DE3) cells containing the respective Cas9 plasmids were grown at 37°C to an OD 600 of 0.6 and protein expression was induced by 1mM IPTG for 16 hr at 18°C.
  • Cells were harvested and lysed by sonication in lysis buffer [50 mM Tris-HCl (pH 7.5), 500 mM NaCl, 5 mM imidazole, 1 mM DTT] supplemented with 1 mg/mL Lysozyme and protease inhibitor cocktail (Sigma).
  • the lysate was then run through a Ni 2+ -NTA agarose column (Qiagen), and the bound protein was eluted with 300 mM imidazole and dialyzed into storage buffer [20 mM HEPES-NaOH (pH 7.5), 250 mM NaCl, 1 mM DTT].
  • storage buffer [20 mM HEPES-NaOH (pH 7.5), 250 mM NaCl, 1 mM DTT].
  • 6xHis-tagged proteins were expressed in E. coli strain BL21 Rosetta (DE3). Cells were grown at 37 qC to an optical density (OD 600 ) of 0.6 in a shaking incubator. The bacterial cultures were cooled to 18°C, and protein expression was induced by adding 1 mM IPTG for overnight expression.
  • a dsDNA target library with randomized PAM sequences was generated by overlapping PCR, with the forward primer containing the 10-nt randomized PAM region.
  • the library was gel-purified and subjected to in vitro cleavage reaction by purified Cas9 along with T7- transcribed sgRNAs.300 nM Cas9:sgRNA complex was used to cleave 300 nM of the target fragment in 1X NEBuffer 3.1 (NEB) at 37 ⁇ C for 1 hr.
  • the reaction was then treated with proteinase K at 50 ⁇ C for 10 minutes and run on a 4% agarose/1xTAE gel.
  • the cleavage product was excised, eluted, and cloned using a previously described protocol (Zhang et al., 2012), with modifications. Briefly, DNA ends were repaired, non-templated 2’-deoxyadenosine tails were added, and Y-shaped adapters were ligated. After PCR, the product was quantitated with KAPA Library Quantification Kit and sequenced using a NextSeq 500 (Illumina) to obtain 75 nt paired- end reads. Sequences were analyzed with custom scripts and R.
  • Lentiviral transduction of K562 cells to stably express Nme2Cas9 K562 cells stably expressing Nme2Cas9 were generated as previously described for Nme1Cas9 (Amrani et al., 2018).
  • the lentiviral vector was co- transfected into HEK293T cells along with the packaging plasmids (Addgene 12260 & 12259) in 6-well plates using TransIT-LT1 transfection reagent (Mirus Bio). After 24 hours, culture media was aspirated from the transfected cells and replaced with 1 mL of fresh DMEM.
  • the supernatant containing the virus was collected and filtered through a 0.45 mm filter.10 uL of the undiluted supernatant along with 2.5 ug of Polybrene was used to transduce ⁇ 10 6 K562 cells in 6-well plates. The transduced cells were selected using media supplemented with 2.5 mg/mL puromycin.
  • the Neon electroporation system was used exactly as described (Amrani et al., 2018). Briefly, 40 picomoles of 3xNLS-Nme2Cas9 along with 50 picomoles of T7-transcribed sgRNA was assembled in buffer R and electroporated using 10 mL Neon tips. After electroporation, cells were plated in pre-warmed 24-well plates containing the appropriate culture media without antibiotics. Electroporation parameters (voltage, width, number of pulses) were 1150 V, 20 ms, 2 pulses for HEK293T cells; 1000 V, 50 ms, 1 pulse for K562 cells.
  • GUIDE-seq experiments were performed as described previously (Tsai et al., 2014), with minor modifications (Bolukbasi et al., 2015a). Briefly, HEK293T cells were transfected with 200 ng of Cas9 plasmid, 200 ng of sgRNA plasmid, and 7.5 pmol of annealed GUIDE-seq oligonucleotides using Polyfect (Qiagen). Alternatively, Hepa1-6 cells were transfected as described above. Genomic DNA was extracted with a DNeasy Blood and Tissue kit (Qiagen) 72 h after transfection according to the manufacturer’s protocol.
  • AAV8.Nme2Cas9+sgRNA delivery and liver tissue processing For the AAV8 vector injections, 8-week-old female C57BL/6NJ mice were injected with 4 x10 11 genome copies per mouse via tail vein, with the sgRNA targeting a validated site in either Pcsk9 or Rosa26. Mice were sacrificed 28 days after vector administration and liver tissues were collected for analysis. Liver tissues were fixed in 4% formalin overnight, embedded in paraffin, sectioned and stained with hematoxylin and eosin (H&E). Blood was drawn from the facial vein at 0, 14 and 28 days post injection, and serum was isolated using a serum separator (BD, Cat. No.365967) and stored at -80°C until assay. Serum cholesterol level was measured using the InfinityTM colorimetric endpoint assay (Thermo-Scientific) following the
  • KSOM Potassium-Supplemented Simplex Optimized Medium, Millipore, Cat. No. MR-106-D
  • 3x10 9 or 3x10 8 GCs of AAV6.Nme2Cas9.sgTyr vector were rinsed in M2 and transferred to fresh KSOM for overnight culture. The next day, the embryos that advanced to 2-cell stage were transferred into the oviduct of pseudopregnant recipients and allowed to develop to term.
  • NmeCas9 is an intrinsically high-fidelity genome editing platform. BioRxiv, https://doi.org/10.1101/172650.
  • CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709-1712.
  • DNA-binding-domain fusions enhance the targeting range and precision of Cas9. Nat. Methods 12, 1150-1156.
  • SpCas9 variant is identified by in vivo screening in yeast. Nat. Biotechnol.36, 265-271.
  • Esvelt, K.M. Mali, P., Braff, J.L., Moosburner, M., Yaung, S.J., and Church, G.M.
  • Promoter traps in embryonic stem cells a genetic screen to identify and mutate developmental genes in mice. Genes Dev.5, 1513-1523.
  • Neisseria meningitidis CRISPR-Cas9 system enables specific genome editing in mammalian cells. Mol. Ther.24, 645-654.
  • GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol.33, 187-197.
  • RNA-Seq Strand-specific libraries for high throughput RNA sequencing
  • CRISPRseek a bioconductor package to identify target-specific guide RNAs for CRISPR-Cas9 genome-editing systems.
  • GUIDEseq a bioconductor package to analyze GUIDE-Seq datasets for CRISPR- Cas nucleases. BMC Genomics 18, 379.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Plant Pathology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Virology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Diabetes (AREA)
  • Veterinary Medicine (AREA)
  • Cell Biology (AREA)
  • Mycology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention is related to the field of gene editing. In particular, the gene editing is directed toward single nucleotide base editing. For example, such single nucleotide base editing results in a conversion of a OG base pair to a T*A base pair. The high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein. The compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein to have a gene editing window that can edit sites that are not targetable by other conventional SpyCas9 base editor platforms.

Description

PROGRAMMABLE DNA BASE EDITING BY NME2CAS9-DEAMINASE
FUSION PROTEINS CROSS-REFERENCE TO RELATED APPLICATION
This application claims priority to U.S. Provisional Patent Application No. 62/745,666, filed October 15, 2018, herein incorporated by reference in it's entirety. Field Of The Invention
The present invention is related to the field of gene editing. In particular, the gene editing is directed toward single nucleotide base editing. For example, such single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair. The high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein. The compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein to have a gene editing window that can edit sites that are not targetable by other conventional SpyCas9 base editor platforms. Background
Many human diseases arise due to the mutation of a single base. The ability to correct such genetic aberrations is paramount in treating these genetic disorders. Clustered regularly interspaced short palindromic repeats (CRISPR) along with CRISPR associated (Cas) proteins comprise an RNA-guided adaptive immune system in archaea and bacteria. These systems provide immunity by targeting and inactivating nucleic acids that originate from foreign genetic elements.
SpyCas9 base editing platforms cannot be used to target all single-base mutations due to their limited editing windows. The editing window is constrained in part by the requirement for an NGG PAM and by the requirement that the edited base(s) be a very precise distance from the PAM. SpyCas9 is also intrinsically associated with high off-targeting effects in genome editing.
What is needed in the art is a highly accurate Cas9 single base editing platform having a programmable target specificity due to recognition of a diverse population of PAM sites. Summary Of The Invention
The present invention is related to the field of gene editing. In particular, the gene editing is directed toward single nucleotide base editing. For example, such single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair. The high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein. The compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein to have a gene editing window that is superior to other conventional SpyCas9 base editor platforms.
In one embodiment, the present invention contemplates a mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N4CC nucleotide sequence. In one embodiment, said protein is Nme2Cas9. In one embodiment, said protein further comprises a nuclear localization signal protein. In one embodiment, said nucleotide deaminase is a cytidine deaminase. In one embodiment, said nucleotide deaminase is an adenosine deaminase. In one embodiment, the protein further comprises a uracil glycosylase inhibitor. In one embodiment, the said nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS. In one embodiment, said binding region is a protospacer accessory motif interacting domain. In one embodiment, said protospacer accessory motif interacting domain comprises said mutation. In one embodiment, said mutation is a D16A mutation. In one embodiment, said mutated NmeCas9 protein further comprises CBE4. In one embodiment, said mutated NmeCas9 protein further comprises a linker. In one embodiment, said linker is a 73aa linker. In one embodiment, said linker is a 3xHA-tag.
In one embodiment, the present invention contemplates a construct, wherein said construct is an optimized nNme2Cas9-ABEmax.
In one embodiment, the present invention contemplates a construct, wherein said construct is a nNme2Cas9-CBE4.
In one embodiment, the present invention contemplates a construct, wherein said construct is a YE1-BE3-nNme2Cas9 (D16A)-UGI.
In one embodiment, the present invention contemplates an adeno-associated virus comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N4CC nucleotide sequence. In one embodiment, said virus is an adeno-associated virus 8. In one embodiment, said virus is an adeno-associated virus 6. In one embodiment, said protein is Nme2Cas9. In one embodiment, said protein further comprises a nuclear localization signal protein. In one embodiment, said nucleotide deaminase is a cytidine deaminase. In one embodiment, said nucleotide deaminase is an adenosine deaminase. In one embodiment, the protein further comprises a uracil glycosylase inhibitor. In one embodiment, the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS. In one embodiment, said binding region is a protospacer accessory motif interacting domain. In one embodiment, said protospacer accessory motif interacting domain comprises said mutation. In one embodiment, said mutation is a D16A mutation. In one embodiment, said mutated NmeCas9 protein further comprises CBE4. In one embodiment, said mutated NmeCas9 protein further comprises a linker. In one embodiment, said linker is a 73aa linker. In one embodiment, said linker is a 3xHA-tag.
In one embodiment, the present invention contemplates a construct, wherein said construct is an optimized nNme2Cas9-ABEmax.
In one embodiment, the present invention contemplates a construct, wherein said construct is a nNme2Cas9-CBE4.
In one embodiment, the present invention contemplates a construct, wherein said construct is a YE1-BE3-nNme2Cas9 (D16A)-UGI.
In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence; ii) a mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence; b) contacting said nucleotide sequence with said mutated NmeCas9 protein under conditions such that said binding region attaches to said N4CC nucleotide sequence; and c) replacing said mutated single base with a wild type base with said mutated NmeCas9 protein. In one embodiment, said protein is Nme2Cas9. In one embodiment, said protein further comprises a nuclear localization signal protein. In one embodiment, said nucleotide deaminase is a cytidine deaminase. In one embodiment, said nucleotide deaminase is an adenosine deaminase. In one embodiment, the protein further comprises a uracil glycosylase inhibitor. In one embodiment, the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS. In one embodiment, said binding region is a protospacer accessory motif interacting domain. In one embodiment, said protospacer accessory motif interacting domain comprises said mutation. In one embodiment, said mutation is a D16A mutation. In one embodiment, said mutated NmeCas9 protein further comprises CBE4. In one embodiment, said mutated NmeCas9 protein further comprises a linker. In one embodiment, said linker is a 73aa linker. In one embodiment, said linker is a 3xHA-tag. In one embodiment, said gene encodes a tyrosinase. In one embodiment, said gene is Fah. In one embodiment, said gene is c-fos.
In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) an adeno-associated virus comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence; b) treating said patient with said adeno- associated virus under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop. In one embodiment, said gene encodes a tyrosinase protein. In one embodiment, said genetically-based medical condition is tyrosinemia. In one embodiment, said virus is an adeno-associated virus 8. In one embodiment, said virus is an adeno-associated virus 6. In one embodiment, said protein is Nme2Cas9. In one embodiment, said protein further comprises a nuclear localization signal protein. In one embodiment, said nucleotide deaminase is a cytidine deaminase. In one embodiment, said nucleotide deaminase is an adenosine deaminase. In one embodiment, the protein further comprises a uracil glycosylase inhibitor. In one embodiment, the nuclear localization signal protein includes, but is not limited to, nucleoplasmin (NLS) and/or SV40 NLS and/or C-myc NLS. In one embodiment, said binding region is a protospacer accessory motif interacting domain. In one embodiment, said protospacer accessory motif interacting domain comprises said mutation. In one embodiment, said mutation is a D16A mutation. In one embodiment, said mutated NmeCas9 protein further comprises CBE4. In one embodiment, said mutated NmeCas9 protein further comprises a linker. In one embodiment, said linker is a 73aa linker. In one embodiment, said linker is a 3xHA-tag. In one embodiment, said gene encodes a tyrosinase. In one embodiment, said gene is Fah. In one embodiment, said gene is c-fos. In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) an optimized nNme2Cas9-ABEmax, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence; b) treating said patient with said optimized nNme2Cas9-ABEmax under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) a nNme2Cas9-CBE4, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence; b) treating said patient with said nNme2Cas9-CBE4 under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
In one embodiment, the present invention contemplates a method, comprising: a) providing; i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition; ii) a YE1-BE3-nNme2Cas9 (D16A)-UGI, comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence; b) treating said patient with said nNme2Cas9-CBE4 under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop. Definitions
To facilitate the understanding of this invention, a number of terms are defined below. Terms defined herein have meanings as commonly understood by a person of ordinary skill in the areas relevant to the present invention. Terms such as“a”,“an” and“the” are not intended to refer to only a singular entity, but include the general class of which a specific example may be used for illustration. The terminology herein is used to describe specific embodiments of the invention, but their usage does not delimit the invention, except as outlined in the claims.
As used herein, the term“edit”“editing” or“edited” refers to a method of altering a nucleic acid sequence of a polynucleotide (e.g., for example, a wild type naturally occurring nucleic acid sequence or a mutated naturally occurring sequence) by selective deletion of a specific genomic target. Such a specific genomic target includes, but is not limited to, a chromosomal region, a gene, a promoter, an open reading frame or any nucleic acid sequence.
As used herein, the term“single base” refers to one, and only one, nucleotide within a nucleic acid sequence. When used in the context of single base editing, it is meant that the base at a specific position within the nucleic acid sequence is replaced with a different base. This replacement may occur by many mechanisms, including but not limited to, substitution or modification.
As used herein, the term“target” or“target site” refers to a pre-identified nucleic acid sequence of any composition and/or length. Such target sites include, but is not limited to, a chromosomal region, a gene, a promoter, an open reading frame or any nucleic acid sequence. In some embodiments, the present invention interrogates these specific genomic target sequences with complementary sequences of gRNA.
The term“on-target binding sequence” as used herein, refers to a subsequence of a specific genomic target that may be completely complementary to a programmable DNA binding domain and/or a single guide RNA sequence.
The term“off-target binding sequence” as used herein, refers to a subsequence of a specific genomic target that may be partially complementary to a programmable DNA binding domain and/or a single guide RNA sequence.
The term“effective amount” as used herein, refers to a particular amount of a
pharmaceutical composition comprising a therapeutic agent that achieves a clinically beneficial result (i.e., for example, a reduction of symptoms). Toxicity and therapeutic efficacy of such compositions can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio LD50/ED50. Compounds that exhibit large therapeutic indices are preferred. The data obtained from these cell culture assays and additional animal studies can be used in formulating a range of dosage for human use. The dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage varies within this range depending upon the dosage form employed, sensitivity of the patient, and the route of administration.
The term“symptom”, as used herein, refers to any subjective or objective evidence of disease or physical disturbance observed by the patient. For example, subjective evidence is usually based upon patient self-reporting and may include, but is not limited to, pain, headache, visual disturbances, nausea and/or vomiting. Alternatively, objective evidence is usually a result of medical testing including, but not limited to, body temperature, complete blood count, lipid panels, thyroid panels, blood pressure, heart rate, electrocardiogram, tissue and/or body imaging scans.
The term“disease” or“medical condition”, as used herein, refers to any impairment of the normal state of the living animal or plant body or one of its parts that interrupts or modifies the performance of the vital functions. Typically manifested by distinguishing signs and symptoms, it is usually a response to: i) environmental factors (as malnutrition, industrial hazards, or climate); ii) specific infective agents (as worms, bacteria, or viruses); iii) inherent defects of the organism (as genetic anomalies); and/or iv) combinations of these factors.
The terms "reduce," "inhibit," "diminish," "suppress," "decrease,"“prevent” and grammatical equivalents (including“lower,”“smaller,” etc.) when in reference to the expression of any symptom in an untreated subject relative to a treated subject, mean that the quantity and/or magnitude of the symptoms in the treated subject is lower than in the untreated subject by any amount that is recognized as clinically relevant by any medically trained personnel. In one embodiment, the quantity and/or magnitude of the symptoms in the treated subject is at least 10% lower than, at least 25% lower than, at least 50% lower than, at least 75% lower than, and/or at least 90% lower than the quantity and/or magnitude of the symptoms in the untreated subject.
The term "attached" as used herein, refers to any interaction between a medium (or carrier) and a drug. Attachment may be reversible or irreversible. Such attachment includes, but is not limited to, covalent bonding, ionic bonding, Van der Waais forces or friction, and the like. A drug is attached to a medium (or carrier) if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc.
The term "drug" or "compound" as used herein, refers to any pharmacologically active substance capable of being administered which achieves a desired effect. Drugs or compounds can be synthetic or naturally occurring, non-peptide, proteins or peptides, oligonucleotides or nucleotides, polysaccharides or sugars.
The term "administered" or "administering", as used herein, refers to any method of providing a composition to a patient such that the composition has its intended effect on the patient. An exemplary method of administering is by a direct mechanism such as, local tissue administration (i.e., for example, extravascular placement), oral ingestion, transdermal patch, topical, inhalation, suppository etc.
The term "patient" or“subject”, as used herein, is a human or animal and need not be hospitalized. For example, out-patients, persons in nursing homes are "patients." A patient may comprise any age of a human or non-human animal and therefore includes both adult and juveniles (i.e., children). It is not intended that the term "patient" connote a need for medical treatment, therefore, a patient may voluntarily or involuntarily be part of experimentation whether clinical or in support of basic science studies.
The term“affinity” as used herein, refers to any attractive force between substances or particles that causes them to enter into and remain in chemical combination. For example, an inhibitor compound that has a high affinity for a receptor will provide greater efficacy in preventing the receptor from interacting with its natural ligands, than an inhibitor with a low affinity.
The term "pharmaceutically" or "pharmacologically acceptable", as used herein, refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
The term, "pharmaceutically acceptable carrier", as used herein, includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, coatings, isotonic and absorption delaying agents, liposome, commercially available cleansers, and the like. Supplementary bioactive ingredients also can be incorporated into such carriers.
The term“viral vector” encompasses any nucleic acid construct derived from a virus genome capable of incorporating heterologous nucleic acid sequences for expression in a host organism. For example, such viral vectors may include, but are not limited to, adeno-associated viral vectors, lentiviral vectors, SV40 viral vectors, retroviral vectors, adenoviral vectors.
Although viral vectors are occasionally created from pathogenic viruses, they may be modified in such a way as to minimize their overall health risk. This usually involves the deletion of a part of the viral genome involved with viral replication. Such a virus can efficiently infect cells but, once the infection has taken place, the virus may require a helper virus to provide the missing proteins for production of new virions. Preferably, viral vectors should have a minimal effect on the physiology of the cell it infects and exhibit genetically stable properties (e.g., do not undergo spontaneous genome rearrangement). Most viral vectors are engineered to infect as wide a range of cell types as possible. Even so, a viral receptor can be modified to target the virus to a specific kind of cell. Viruses modified in this manner are said to be pseudotyped. Viral vectors are often engineered to incorporate certain genes that help identify which cells took up the viral genes. These genes are called marker genes. For example, a common marker gene confers antibiotic resistance to a certain antibiotic.
As used herein the“ROSA26 gene” or“Rosa26 gene” refers to a human or mouse (respectively) locus that is widely used for achieving generalized expression in the mouse.
Targeting to the ROSA26 locus may be achieved by introducing a desired gene into the first intron of the locus, at a unique XbaI site approximately 248 bp upstream of the original gene trap line. A construct may be constructed using an adenovirus splice acceptor followed by a gene of interest and a polyadenylation site inserted at the unique XbaI site. A neomycin resistance cassette may also be included in the targeting vector.
As used herein the“PCSK9 gene” or“Pcsk9 gene” refers to a human or mouse
(respectively) locus that encodes a PCSK9 protein. The PCSK9 gene resides on chromosome 1 at the band 1p32.3 and includes 13 exons. This gene may produce at least two isoforms through alternative splicing.
The term“proprotein convertase subtilisin/kexin type 9” and“PCSK9” refers to a protein encoded by a gene that modulates low density lipoprotein levels. Proprotein convertase subtilisin/kexin type 9, also known as PCSK9, is an enzyme that in humans is encoded by the PCSK9 gene. Seidah et al., "The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation" Proc. Natl. Acad. Sci. U.S.A.100 (3): 928–933 (2003). Similar genes (orthologs) are found across many species. Many enzymes, including PSCK9, are inactive when they are first synthesized, because they have a section of peptide chains that blocks their activity; proprotein convertases remove that section to activate the enzyme. PSCK9 is believed to play a regulatory role in cholesterol homeostasis. For example, PCSK9 can bind to the epidermal growth factor-like repeat A (EGF-A) domain of the low-density lipoprotein receptor (LDL-R) resulting in LDL-R internalization and degradation. Clearly, it would be expected that reduced LDL-R levels result in decreased metabolism of LDL- C, which could lead to hypercholesterolemia.
The term“hypercholesterolemia” as used herein, refers to any medical condition wherein blood cholesterol levels are elevated above the clinically recommended levels. For example, if cholesterol is measured using low density lipoproteins (LDLs), hypercholesterolemia may exist if the measured LDL levels are above, for example, approximately 70 mg/dl. Alternatively, if cholesterol is measured using free plasma cholesterol, hypercholesterolemia may exist if the measured free cholesterol levels are above, for example, approximately 200-220 mg/dl.
As used herein, the term“CRISPRs” or“Clustered Regularly Interspaced Short
Palindromic Repeats” refers to an acronym for DNA loci that contain multiple, short, direct repetitions of base sequences. Each repetition contains a series of bases followed by 30 or so base pairs known as "spacer DNA". The spacers are short segments of DNA from a virus and may serve as a 'memory' of past exposures to facilitate an adaptive defense against future invasions.
As used herein, the term“Cas” or“CRISPR-associated (cas)” refers to genes often associated with CRISPR repeat-spacer arrays.
As used herein, the term“Cas9” refers to a nuclease from Type II CRISPR systems, an enzyme specialized for generating double-strand breaks in DNA, with two active cutting sites (the HNH and RuvC domains), one for each strand of the double helix. Jinek combined tracrRNA and spacer RNA into a "single-guide RNA" (sgRNA) molecule that, mixed with Cas9, could find and cleave DNA targets through Watson-Crick pairing between the guide sequence within the sgRNA and the target DNA sequence. The term“protospacer adjacent motif” (or PAM) as used herein, refers to a DNA sequence that may be required for a Cas9/sgRNA to form an R-loop to interrogate a specific DNA sequence through Watson-Crick pairing of its guide RNA with the genome. The PAM specificity may be a function of the DNA-binding specificity of the Cas9 protein (e.g., a “protospacer adjacent motif recognition domain” at the C-terminus of Cas9).
As used herein, the term“sgRNA” refers to single guide RNA used in conjunction with CRISPR associated systems (Cas). sgRNAs are a fusion of crRNA and tracrRNA and contain nucleotides of sequence complementary to the desired target site. Jinek et al.,“A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity” Science 337(6096):816- 821 (2012) Watson-Crick pairing of the sgRNA with the target site permits R-loop formation, which in conjunction with a functional PAM permits DNA cleavage or in the case of nuclease- deficient Cas9 allows binds to the DNA at that locus.
As used herein, the term“fluorescent protein” refers to a protein domain that comprises at least one organic compound moiety that emits fluorescent light in response to the appropriate wavelengths. For example, fluorescent proteins may emit red, blue and/or green light. Such proteins are readily commercially available including, but not limited to: i) mCherry (Clonetech Laboratories): excitation: 556/20 nm (wavelength/bandwidth); emission: 630/91 nm; ii) sfGFP (Invitrogen): excitation: 470/28 nm; emission: 512/23 nm; iii) TagBFP (Evrogen): excitation 387/11 nm; emission 464/23 nm.
As used herein, the term“sgRNA” refers to single guide RNA used in conjunction with CRISPR associated systems (Cas). sgRNAs contains nucleotides of sequence complementary to the desired target site. Watson-crick pairing of the sgRNA with the target site recruits the nuclease-deficient Cas9 to bind the DNA at that locus.
As used herein, the term“orthogonal” refers targets that are non-overlapping, uncorrelated, or independent. For example, if two orthogonal nuclease-deficient Cas9 gene fused to different effector domains were implemented, the sgRNAs coded for each would not cross- talk or overlap. Not all nuclease-deficient Cas9 genes operate the same, which enables the use of orthogonal nuclease-deficient Cas9 gene fused to a different effector domains provided the appropriate orthogonal sgRNAs.
As used herein, the term“phenotypic change” or“phenotype” refers to the composite of an organism's observable characteristics or traits, such as its morphology, development, biochemical or physiological properties, phenology, behavior, and products of behavior.
Phenotypes result from the expression of an organism's genes as well as the influence of environmental factors and the interactions between the two.
"Nucleic acid sequence" and "nucleotide sequence" as used herein refer to an
oligonucleotide or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin which may be single- or double-stranded, and represent the sense or antisense strand.
The term "an isolated nucleic acid”, as used herein, refers to any nucleic acid molecule that has been removed from its natural state (e.g., removed from a cell and is, in a preferred embodiment, free of other genomic nucleic acid).
The terms "amino acid sequence" and "polypeptide sequence" as used herein, are interchangeable and to refer to a sequence of amino acids.
As used herein the term "portion" when in reference to a protein (as in "a portion of a given protein") refers to fragments of that protein. The fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.
The term "portion" when used in reference to a nucleotide sequence refers to fragments of that nucleotide sequence. The fragments may range in size from 5 nucleotide residues to the entire nucleotide sequence minus one nucleic acid residue.
As used herein, the terms "complementary" or "complementarity" are used in reference to "polynucleotides" and "oligonucleotides" (which are interchangeable terms that refer to a sequence of nucleotides) related by the base-pairing rules. For example, the sequence "C-A-G- T," is complementary to the sequence "G-T-C-A." Complementarity can be "partial" or "total." "Partial" complementarity is where one or more nucleic acid bases is not matched according to the base pairing rules. "Total" or "complete" complementarity between nucleic acids is where each and every nucleic acid base is matched with another base under the base pairing rules. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.
The terms "homology" and "homologous" as used herein in reference to nucleotide sequences refer to a degree of complementarity with other nucleotide sequences. There may be partial homology or complete homology (i.e., identity). A nucleotide sequence which is partially complementary, i.e., "substantially homologous," to a nucleic acid sequence is one that at least partially inhibits a completely complementary sequence from hybridizing to a target nucleic acid sequence. The inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or Northern blot, solution hybridization and the like) under conditions of low stringency. A substantially homologous sequence or probe will compete for and inhibit the binding (i.e., the hybridization) of a completely homologous sequence to a target sequence under conditions of low stringency. This is not to say that conditions of low stringency are such that non-specific binding is permitted; low stringency conditions require that the binding of two sequences to one another be a specific (i.e., selective) interaction. The absence of non-specific binding may be tested by the use of a second target sequence which lacks even a partial degree of complementarity (e.g., less than about 30% identity); in the absence of non-specific binding the probe will not hybridize to the second non-complementary target.
The terms“homology” and“homologous” as used herein in reference to amino acid sequences refer to the degree of identity of the primary structure between two amino acid sequences. Such a degree of identity may be directed to a portion of each amino acid sequence, or to the entire length of the amino acid sequence. Two or more amino acid sequences that are “substantially homologous” may have at least 50% identity, preferably at least 75% identity, more preferably at least 85% identity, most preferably at least 95%, or 100% identity.
An oligonucleotide sequence which is a "homolog" is defined herein as an
oligonucleotide sequence which exhibits greater than or equal to 50% identity to a sequence, when sequences having a length of 100 bp or larger are compared.
Low stringency conditions comprise conditions equivalent to binding or hybridization at 42°C in a solution consisting of 5 x SSPE (43.8 g/l NaCl, 6.9 g/l NaH2PO4·H2O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5x Denhardt's reagent {50x Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; Sigma)} and 100 mg/ml denatured salmon sperm DNA followed by washing in a solution comprising 5x SSPE, 0.1% SDS at 42°C when a probe of about 500 nucleotides in length is employed. Numerous equivalent conditions may also be employed to comprise low stringency conditions; factors such as the length and nature (DNA, RNA, base composition) of the probe and nature of the target ( DNA, RNA, base composition, present in solution or immobilized, etc.) and the concentration of the salts and other components (e.g., the presence or absence of formamide, dextran sulfate, polyethylene glycol), as well as components of the hybridization solution may be varied to generate conditions of low stringency hybridization different from, but equivalent to, the above listed conditions. In addition, conditions which promote hybridization under conditions of high stringency (e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc.) may also be used.
As used herein, the term "hybridization" is used in reference to the pairing of
complementary nucleic acids using any process by which a strand of nucleic acid joins with a complementary strand through base pairing to form a hybridization complex. Hybridization and the strength of hybridization (i.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementarity between the nucleic acids, stringency of the conditions involved, the Tm of the formed hybrid, and the G:C ratio within the nucleic acids.
As used herein the term "hybridization complex" refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bounds between complementary G and C bases and between complementary A and T bases; these hydrogen bonds may be further stabilized by base stacking interactions. The two complementary nucleic acid sequences hydrogen bond in an antiparallel configuration. A hybridization complex may be formed in solution (e.g., C0 t or R0 t analysis) or between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized to a solid support (e.g., a nylon membrane or a nitrocellulose filter as employed in Southern and Northern blotting, dot blotting or a glass slide as employed in in situ hybridization, including FISH (fluorescent in situ hybridization)).
DNA molecules are said to have "5' ends" and "3' ends" because mononucleotides are reacted to make oligonucleotides in a manner such that the 5' phosphate of one mononucleotide pentose ring is attached to the 3' oxygen of its neighbor in one direction via a phosphodiester linkage. Therefore, an end of an oligonucleotide is referred to as the "5' end" if its 5' phosphate is not linked to the 3' oxygen of a mononucleotide pentose ring. An end of an oligonucleotide is referred to as the "3' end" if its 3' oxygen is not linked to a 5' phosphate of another
mononucleotide pentose ring. As used herein, a nucleic acid sequence, even if internal to a larger oligonucleotide, also may be said to have 5' and 3' ends. In either a linear or circular DNA molecule, discrete elements are referred to as being "upstream" or 5' of the "downstream" or 3' elements. This terminology reflects the fact that transcription proceeds in a 5' to 3' fashion along the DNA strand. The promoter and enhancer elements which direct transcription of a linked gene are generally located 5' or upstream of the coding region. However, enhancer elements can exert their effect even when located 3' of the promoter element and the coding region. Transcription termination and polyadenylation signals are located 3' or downstream of the coding region.
The term "transfection" or "transfected" refers to the introduction of foreign DNA into a cell.
As used herein, the terms "nucleic acid molecule encoding", "DNA sequence encoding," and "DNA encoding" refer to the order or sequence of deoxyribonucleotides along a strand of deoxyribonucleic acid. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide (protein) chain. The DNA sequence thus codes for the amino acid sequence.
As used herein, the term "gene" means the deoxyribonucleotide sequences comprising the coding region of a structural gene and including sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of about 1 kb on either end such that the gene corresponds to the length of the full-length mRNA. The sequences which are located 5' of the coding region and which are present on the mRNA are referred to as 5' non-translated sequences. The sequences which are located 3' or downstream of the coding region and which are present on the mRNA are referred to as 3' non-translated sequences. The term "gene" encompasses both cDNA and genomic forms of a gene. A genomic form or clone of a gene contains the coding region interrupted with non-coding sequences termed "introns" or "intervening regions" or "intervening sequences." Introns are segments of a gene which are transcribed into
heterogeneous nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers. Introns are removed or "spliced out" from the nuclear or primary transcript; introns therefore are absent in the messenger RNA (mRNA) transcript. The mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide.
In addition to containing introns, genomic forms of a gene may also include sequences located on both the 5' and 3' end of the sequences which are present on the RNA transcript.
These sequences are referred to as "flanking" sequences or regions (these flanking sequences are located 5' or 3' to the non-translated sequences present on the mRNA transcript). The 5' flanking region may contain regulatory sequences such as promoters and enhancers which control or influence the transcription of the gene. The 3' flanking region may contain sequences which direct the termination of transcription, posttranscriptional cleavage and polyadenylation.
The term "label" or "detectable label" are used herein, to refer to any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Such labels include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., Dynabeads®), fluorescent dyes (e.g., fluorescein, texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3H, 125I, 35S, 14C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and calorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Patents teaching the use of such labels include, but are not limited to, U.S. Pat. Nos.3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241 (all herein incorporated by reference in their entirety). The labels contemplated in the present invention may be detected by many methods. For example, radiolabels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted light. Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting, the reaction product produced by the action of the enzyme on the substrate, and calorimetric labels are detected by simply visualizing the colored label. Brief Description Of The Drawings
The patent or application file contains at least one drawing executed in color. Copies of this patent or patent application publication with color drawing(s) will be provided by the Office upon request and payment of the necessary fee.
Figure 1 illustrates exemplary schematic embodiments of an NmeCas9 deaminase fusion protein single base editor and exemplary constructed plasmids of base editors.
Figure 1A shows an exemplary YE1-BE3-nNme2Cas9 (D16A)-UGI construct.
Figure 1B shows an exemplary ABE7.10 nNme2Cas9 (D16A) construct.
Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct comprising two SV40 NLS sequences. Figure 1D shows an exemplary nNme2Cas9-CBE4 (also called a BE4-nNme2Cas9
(D16A)-UGI-UGI) construct.
Figure 1E shows an exemplary optimized nNme2Cas9-ABEmax construct.
Figure 2 presents exemplary data of the electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3-nNme2Cas9 (D16A)-UGI fusion protein efficiently converting C to T at endogenous target site 25 (TS25) in HEK293T cells via nucleofection.
Figure 2A shows exemplary sequences for a TS25 endogenous target site (within the black rectangle). GN23 sgRNA base-pairs with the target DNA strand, leaving the displaced DNA strand for cytidine deaminase to edit (e.g. new green nucleotides). Figure 2B shows exemplary sequencing data showing a doublet nucleotide peak (7th position from 5’ end; arrow) demonstrating the successful single base editing of a cytidine to a thymidine (e.g., a C•G base pair conversion to a T•A base pair). Figure 2C shows an exemplary quantitation of the data shown in Figure 2B plotting the percent conversion of C ® T single base editing. The percentage of C converted to T is about 40% in the base editor- and sgRNA-treated sample (p-value = 6.88 x 10-6). The“no sgRNA” control displays the background noise due to Sanger sequencing. EditR (Kluesner et al., 2018) was used to perform the analysis.
Figure 3 presents exemplary specific UGI target sites that were respectively integrated into YE1-BE3-nNme2Cas9/D16A mutant fusion proteins and co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line. Converted bases are highlighted in orange color. Background signals were filtered using negative control samples (YE1-BE3- nNme2Cas9 nucleofected K562 cells without sgRNA constructs). N4CC PAMs are boxed. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column.
Figure 3A shows an exemplary EGFP-Site 1.
Figure 3B shows an exemplary EGFP-Site 2.
Figure 3C shows an exemplary EGFP-Site 3.
Figure 3D shows an exemplary EGFP-Site 4.
Figure 3E shows an exemplary deep-sequencing analysis indicating where YE1-BE3- nNme2Cas9 converts C residues to T residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange or yellow color. Background signals were filtered using negative control samples. The highest percentage of editing is 32.50%.
Figure 3F shows an exemplary deep-sequencing analysis indicating where ABE7.10- nNme2Cas9 or ABEmax (Koblan et al., 2018)-nNme2Cas9 converts A residues to G residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange color. Background signals were filtered using negative control samples. The percentage of editing is 0.53% by ABE7.10-nNme2Cas9 or 2.33% by ABEmax-nNme2Cas9 (D16A). Figure 4 presents an exemplary alignment of the wildtype Fah gene with the tyrosinemia Fah mutant gene showing an A-G single base gene editing target site (position 9). The respective SpyCas9 single PAM site and NmeCas9 double PAM sites are indicated for demonstrating the suboptimal targeting window relative to the SpyCas9 PAM site.
Figure 5 illustrates exemplary three closely related Neisseria meningitidis Cas9 orthologs that have distinct PAMs.
Figure 5A shows an exemplary schematic showing mutated residues (orange spheres) between Nme2Cas9 (left) and Nme3Cas9 (right) mapped onto the predicted structure of Nme1Cas9, revealing the cluster of mutations in the PID (black). Figure 5B shows an exemplary experimental workflow of the in vitro PAM discovery assay with a 10-bp randomized PAM region. Following in vitro digestion, adapters were ligated to cleaved products for library construction and sequencing. Figure 5C shows exemplary sequence logos resulting from in vitro PAM discovery reveal the enrichment of a N4GATT PAM for Nme1Cas9, consistent with its previously established specificity.
Figure 5D shows exemplary sequence logos indicating that Nme1Cas9 with its PID
swapped with that of Nme2Cas9 (left) or Nme3Cas9 (right) requires a C at PAM position 5. The remaining nucleotides were not determined with high confidence due to the modest cleavage efficiency of the PID-swapped protein chimeras (see Figure 6C). Figure 5E shows an exemplary sequence logo showing that full-length Nme2Cas9 recognizes an N4CC PAM, based on efficient substrate cleavage of a target pool with a fixed C at PAM position 5, and with PAM nts 1-4 and 6-8 randomized. Figure 6 presents a characterization of Neisseria meningitidis Cas9 orthologs with rapidly-evolving PIDs, as related to Figure 5.
Figure 6A shows an exemplary unrooted phylogenetic tree of NmeCas9 orthologs that are >80% identical to Nme1Cas9. Three distinct branches emerged, with the majority of mutations clustered in the PID. Groups 1 (blue), 2 (orange), and 3 (green) have PIDs with >98%, ~52%, and ~86% identity to Nme1Cas9, respectively. Three representative Cas9 orthologs (one from each group)
(Nme1Cas9, Nme2Cas9 and Nme3Cas9) are indicated.
Figure 6B shows an exemplary schematic showing the CRISPR-cas loci of the strains encoding the three Cas9 orthologs (Nme1Cas9, Nme2Cas9, and Nme3Cas9) from (A). Percent identities of each CRISPR-Cas component with N. meningitidis 8013 (encoding Nme1Cas9) are shown. Blue and red arrows denote pre-crRNA and tracrRNA transcription initiation sites, respectively.
Figure 6C shows an exemplary normalized read counts (% of total reads) from cleaved DNAs from the in vitro assays for intact Nme1Cas9 (grey), for chimeras with Nme1Cas9’s PID swapped with those of Nme2Cas9 and Nme3Cas9 (mixed colors), and for full-length Nme2Cas9 (orange), are plotted. The reduced normalized read counts indicate lower cleavage efficiencies in the chimeras. Figure 6D shows an exemplary sequence logos from the in vitro PAM discovery assay on an NNNNCNNN PAM pool by Nme1Cas9 with its PID swapped with those of Nme2Cas9 (left) or Nme3Cas9 (right).
Figure 7 presents exemplary data showing that Nme2Cas9 uses a 22-24 nt spacer to edit sites adjacent to an N4CC PAM. All experiments were done in triplicate, and error bars represent the standard error of the mean (s.e.m.).
Figure 7A shows an exemplary schematic diagram depicting transient transfection and editing of HEK293T TLR2.0 cells, with mCherry+ cells detected by flow cytometry 72 hours after transfection. Figure 7B shows an exemplary Nme2Cas9 editing of the TLR2.0 reporter. Sites with N4CC PAMs were targeted with varying efficiencies, while no Nme2Cas9 targeting was observed at an N4GATT PAM or in the absence of sgRNA.
SpyCas9 (targeting a previously validated site with an NGG PAM) and
Nme1Cas9 (targeting N4GATT) were used as positive controls.
Figure 7C shows an exemplary effect of spacer length on the efficiency of Nme2Cas9 editing. An sgRNA targeting a single TLR2.0 site, with spacer lengths varying from 24 to 20 nts (including the 5’-terminal G required by the U6 promoter), indicate that highest editing efficiencies are obtained with 22-24 nt spacers.
Figure 7D shows an exemplary An Nme2Cas9 dual nickase can be used in tandem to generate NHEJ- and HDR-based edits in TLR2.0. Nme2Cas9- and sgRNA- expressing plasmids, along with an 800-bp dsDNA donor for homologous repair, were electroporated into HEK293T TLR2.0 cells, and both NHEJ (mCherry+) and HDR (GFP+) outcomes were scored by flow cytometry. HNH nickase,
Nme2Cas9D16A; RuvC nickase, Nme2Cas9H588A. Cleavage sites 32 bp and 64 bp apart were targeted using either nickase. The HNH nickase (Nme2Cas9D16A) yielded efficient editing, particularly with the cleavage sites that were separated by 32 bp, whereas the RuvC nickase (Nme2Cas9H588A) was not effective.
Wildtype Nme2Cas9 was used as a control.
Figure 8 presents exemplary data showing PAM, spacer, and seed requirements for Nme2Cas9 targeting in mammalian cells, as related to Figure 7. All experiments were done in triplicate and error bars represent s.e.m.
Figure 8A shows an exemplary Nme2Cas9 targeting at N4CD sites in TLR2.0, with
editing estimated based on mCherry+ cells. Four sites for each non-C nucleotide at the tested position (N4CA, N4CT and N4CG) were examined, and an N4CC site was used as a positive control.
Figure 8B shows an exemplary Nme2Cas9 targeting at N4DC sites in TLR2.0 [similar to (A)].
Figure 8C shows exemplary guide truncations on a TLR2.0 site (distinct from that in Figure 2C) with a N4CCA PAM, revealing similar length requirements as those observed at the other site. Figure 8D shows exemplary Nme2Cas9 targeting efficiency is differentially sensitive to single-nucleotide mismatches in the seed region of the sgRNA. Data show the effects of walking single-nucleotide sgRNA mismatches along the 23-nt spacer in a TLR2.0 target site.
Figure 9 presents exemplary data showing Nme2Cas9 genome editing at endogenous loci in mammalian cells via multiple delivery methods. All results represent 3 independent biological replicates, and error bars represent s.e.m.
Figure 9A shows an exemplary Nme2Cas9 genome editing of endogenous human sites in HEK293T cells following transient transfection of Nme2Cas9- and sgRNA- expressing plasmids.40 sites were screened initially (Table 1); the 14 sites shown (selected to include representatives of varying editing efficiencies, as measured by TIDE) were then re-analyzed in triplicate. An Nme1Cas9 target site (with an N4GATT PAM) was used as a negative control.
Figure 9B shows exemplary data charts: Left panel: Transient transfection of a single plasmid expressing both Nme2Cas9 and sgRNA (targeting the Pcsk9 and Rosa26 loci) enables editing in Hepa1-6 mouse cells, as detected by TIDE. Right panel: Electroporation of sgRNA plasmids into K562 cells stably expressing Nme2Cas9 from a lentivector results in efficient indel formation.
Figure 9C shows exemplary Nme2Cas9 can be electroporated as an RNP complex to induce genome editing.40 picomoles Cas9 along with 50 picomoles of in vitro- transcribed sgRNAs targeting three different loci were electroporated into HEK293T cells. Indels were measured after 72h using TIDE.
Figure 10 presents exemplary data showing dose dependence and segmental deletions by Nme2Cas9, as related to Figure 9.
Figure 10A shows exemplary increasing the dose of electroporated Nme2Cas9 plasmid (500 ng, vs.200 ng in Figure 3A) improves editing efficiency at two sites (TS16 and TS6). Data provided in yellow are re-used from Figure 9A.
Figure 10B shows exemplary Nme2Cas9 can be used to create precise segmental
deletions. Two TLR2.0 targets with cleavage sites 32 bp apart were targeted simultaneously with Nme2Cas9. The majority of lesions created were deletions of exactly 32 bp (blue). Figure 11 presents exemplary data showing that Nme2Cas9 is subject to inhibition by a subset of type II-C anti-CRISPR families in vitro and in cells. All experiments were done in triplicate and error bars represent s.e.m.
Figure 11A shows exemplary In vitro cleavage assay of Nme1Cas9 and Nme2Cas9 in the presence of five previously characterized anti-CRISPR proteins (10:1 ratio of Acr:Cas9). Top: Nme1Cas9 efficiently cleaves a fragment containing a protospacer with an N4GATT PAM in the absence of an Acr or in the presence of a negative control Acr (AcrE2). All five previously characterized type II-C Acr families inhibited Nme1Cas9, as expected. Bottom: Nme2Cas9 inhibition mirrors that of Nme1Cas9, except for the lack of inhibition by AcrIIC5Smu.
Figure 11B shows exemplary genome editing in the presence of the five previously
described anti-CRISPR families. Plasmids expressing Nme2Cas9 (200 ng), sgRNA (100 ng) and each respective Acr (200 ng) were co-transfected into HEK293T cells, and genome editing was measured using Tracking of Indels by Decompostion (TIDE) 72 hr post transfection. Consistent with our in vitro analyses, all type II-C anti-CRISPRs except AcrIIC5Smu inhibited genome editing, albeit with different efficiencies.
Figure 11C shows exemplary Acr inhibition of Nme2Cas9 is dose-dependent with
distinct apparent potencies. Nme2Cas9 is fully inhibited by AcrIIC1Nme and AcrIIC4Hpa at 2:1 and 1:1 mass ratios of cotransfected Acr and Nme2Cas9 plasmids, respectively.
Figure 12 presents exemplary data showing that a Nme2Cas9 PID swap renders
Nme1Cas9 insensitive to AcrIIC5Smu inhibition, as related to Figure 11. In vitro cleavage by the Nme1Cas9-Nme2Cas9PID chimera in the presence of previously characterized Acr proteins (10 uM Cas9-sgRNA + 100 uM Acr).
Figure 13 presents exemplary data showing orthogonality and relative accuracy of Nme2Cas9 and SpyCas9 at dual target sites, as related to Figure 12.
Figure 13A shows exemplary Nme2Cas9 and SpyCas9 guides are orthogonal. TIDE
results show the frequencies of indels created by both nucleases targeting DS2 with either their cognate sgRNAs or with the sgRNAs of the other ortholog. Figure 13B shows exemplary Nme2Cas9 and SpyCas9 exhibiting comparable on-target editing efficiencies as assessed by GUIDE-seq. Bars indicate on-target read counts from GUIDE-Seq at the three dual sites targeted by each ortholog. Orange bars represent Nme2Cas9 and black bars represent SpyCas9.
Figure 13C shows an exemplary SpyCas9’s on-target vs. off-target read counts for each site. Orange bars represent the on-target reads while black bars represent off- targets.
Figure 13D shows exemplary Nme2Cas9’s on-target vs. off-target reads for each site. Figure 13E bar graphs showing exemplary indel efficiencies (measured by TIDE) at potential off-target sites predicted by CRISPRSeek. On- and off-target site sequences are shown on the left, with the PAM region underlined and sgRNA mismatches and non-consensus PAM nucleotides given in red.
Figure 14 presents exemplary data showing that Nme2Cas9 exhibits little or no detectable off-targeting in mammalian cells.
Figure 14Ashows an exemplary schematic depicting dual sites (DSs) targetable by both SpyCas9 and Nme2Cas9 by virtue of their non-overlapping PAMs. The
Nme2Cas9 PAM (orange) and SpyCas9 PAM (blue) are highlighted. A 24nt Nme2Cas9 guide sequence is indicated in yellow; the corresponding guide sequence for SpyCas9 would be 4nt shorter at the 5’ end.
Figure 14B shows an exemplary Nme2Cas9 and SpyCas9 that both induce indels at DSs.
Six DSs in VEGFA (with GN3GN19NGGNCC sequences) were selected for direct comparisons of editing by the two orthologs. Plasmids expressing each Cas9 (with the same promoter, linkers, tags and NLSs) and its cognate guide were transfected into HEK293T cells. Indel efficiencies were determined by TIDE 72 hrs post transfection. Nme2Cas9 editing was detectable at all six sites and was marginally or significantly more efficient than SpyCas9 at two sites (DS2 and DS6, respectively). SpyCas9 edited four out of the six sites (DS1, DS2, DS4 and DS6), with two sites showing significantly higher editing efficiencies than Nme2Cas9 (DS1 and DS4). DS2, DS4 and DS6 were selected for GUIDE-Seq analysis as Nme2Cas9 was equally efficient, less efficient and more efficient than SpyCas9, respectively, at these sites. Figure 14C shows exemplary Nme2Cas9 genome editing that is highly accurate in human cells. Numbers of off-target sites detected by GUIDE-Seq for each nuclease at individual target sites are shown. In addition to dual sites, we analyzed TS6 (because of its high on-target editing efficiency) and Pcsk9 and Rosa26 sites in mouse Hepa1-6 cells (to measure accuracy in another cell type). Figure 14D shows an exemplary targeted deep sequencing to detect indels in edited cells confirms the high Nme2Cas9 accuracy indicated by GUIDE-seq.
Figure 14E shows an exemplary sequence for the validated off-target site of the Rosa26 guide, showing the PAM region (underlined), the consensus CC PAM dinucleotide (bold), and three mismatches in the PAM-distal portion of the spacer (red).
Figure 15 presents exemplary data showing Nme2Cas9 genome editing in vivo via all-in- one AAV delivery.
Figure 15A shows exemplary workflow for delivery of AAV8.sgRNA.Nme2Cas9 to lower cholesterol levels in mice by targeting Pcsk9. Top: schematic of the all-in- one AAV vector expressing Nme2Cas9 and the sgRNA (individual genome elements not to scale). BGH, bovine growth hormone poly(A) site; HA, epitope tag; NLS, nuclear localization sequence; h, human-codon-optimized. Bottom: Timeline for AAV8.sgRNA.Nme2Cas9 tail-vein injections (4 x 1011 GCs), followed by cholesterol measurements at day 14 and indel, histology and cholesterol analyses at day 28 post-injection.
Figure 15B shows an exemplary TIDE analysis to measure indels in DNA extracted from livers of mice injected with AAV8.Nme2Cas9+sgRNA targeting Pcsk9 and Rosa26 (control) loci. Indel efficiency at the lone off-target site identified by GUIDE-seq for these two sgRNAs (Rosa26|OT1) were also assessed by TIDE. Figure 15C shows an exemplary reduced serum cholesterol levels in mice injected with the Pcsk9-targeting guide compared to the Rosa26-targeting controls. P values are calculated by unpaired two-tailed t-test.
Figure 16 presents exemplary data showing PCSK9 knockdown and liver histology following Nme2Cas9 AAV delivery and editing, related to Figure 15. Figure 16A shows exemplary Western blotting using anti-PCSK9 antibody reveals strongly reduced levels of PCSK9 in the livers of mice treated with sgPcsk9, compared to mice treated with sgRosa26.2ng of recombinant PCSK9 was used as a mobility standard (left-most lane), and a cross-reacting band in the liver samples is indicated by an asterisk. GAPDH was used as loading control (bottom panel). Figure 16B shows exemplary H&E staining from livers of mice injected with
AAV8.Nme2Cas9+sgRosa26 (left) or AAV8.Nme2Cas9+sgPcsk9 (right) vectors. Scale bars, 25 mm.
Figure 17 presents exemplary data showing Tyr editing ex vivo in mouse zygotes, related to Figure 16.
Figure 17A shows an exemplary two sites in Tyr, each with N4CC PAMs, were tested for editing in Hepa1-6 cells. The sgTyr2 guide exhibited higher editing efficiency and was selected for further testing.
Figure 17B shows an exemplary seven mice that survived post-natal development, and each exhibited coat color phenotypes as well as on-target editing, as assayed by TIDE.
Figure 17C shows an exemplary Indel spectra from tail DNA of each mouse from (B), as well as an unedited C57BL/6NJ mouse, as indicated by TIDE analysis.
Efficiencies of insertions (positive) and deletions (negative) of various sizes are indicated.
Figure 18 presents exemplary data showing Nme2Cas9 genome editing ex vivo via all-in- one AAV delivery.
Figure 18A shows an exemplary workflow for single-AAV Nme2Cas9 editing ex vivo to generate albino C57BL/6NJ mice by targeting the Tyr gene. Zygotes are cultured in KSOM containing AAV6.Nme2Cas9:sgTyr for 5-6 hours, rinsed in M2, and cultured for a day before being transferred to the oviduct of pseudo-pregnant recipients.
Figure 18B shows exemplary albino (left) and chinchilla or variegated (middle) mice generated by 3x109 GCs, and chinchilla or variegated mice (right) generated by 3x108 GCs of zygotes with AAV6.Nme2Cas9:sgTyr.
Figure 18C shows an exemplary summary of Nme2Cas9.sgTyr single-AAV ex vivo Tyr editing experiments at two AAV doses. Figure 19 shows an exemplary mCherry reporter assay for nSpCas9-ABEmax and optimized ABEmax-nNme2Cas9 (D16A) activities.
Figure 19A shows exemplary sequence information of sequence information of ABE- mCherry reporter. There is a TAG stop codon in mCherry coding region. In the reporter- integrated stable cell line, there is no mCherry signal. The mCherry signal will show up if the nSpCas9-ABEmax or optimized ABEmax-nNme2Cas9 (D16A) can convert TAG to CAG (which is encoded Gln).
Figure 19B shows an exemplary mCherry signals light up since SpCas9-ABE or
ABEmax-nNme2Cas9 (D16A) is active in the specific region of the mCherry reporter. Upper panel is the negative control, middle panel shows the mCherry signals light up in reporter cells treated with nSpCas9-ABEmax, bottom panel shows the mCherry signals light up in reporter cells treated with optimized ABEmax-nNme2Cas9 (D16A).
Figure 19C shows an exemplary FACs Quantitation of base editing events in mCherry reporter cells transfected with the SpCas9-ABE or ABEmax-nNme2Cas9 (D16A).
N = 6; error bars represent S.D. Results are from biological replicates performed in technical duplicates.
Figure 20 shows an exemplary GFP reporter assay for nSpCas9-CBE4 (Addgene
#100802) and CBE4-nNme2Cas9 (D16A)-UGI-UGI (CBE4 was cloned from Addgene #100802) activities.
Figure 20A shows exemplary sequence information of CBE-GFP reporter. There is a mutation in the fluorophore core region of the GFP reporter line, which converts GYG to GHG. Therefore, there is no GFP signal. The GFP signal will show up if the nSpCas9- CBE4 or CBE4-nNme2Cas9 (D16A)-UGI-UGI can convert CAC to TAC/TAT
(Histidine to Tyrosine).
Figure 20B shows an exemplary GFP signal (green) since nSpCas9-CBE4 or CBE4- nNme2Cas9 (D16A)-UGI-UGI is active in the specific region of the GFP reporter. Upper panel is the negative control. Middle panel shows that the mCherry signals light up in the reporter cells treated with CBE4-nNme2Cas9 (D16A)-UGI-UGI. Bottom panel shows that the GFP signals light up in the reporter cells treated with CBE4-nNme2Cas9 (D16A)-UGI-UGI). Figure 20C shows an exemplary FACs Quantitation of base editing events in GFP reporter cells transfected with nSpCas9-CBE4 or CBE4-nNme2Cas9 (D16A)-UGI-UGI. N = 6; error bars represent S.D. Results are from biological replicates performed in technical duplicates.
Figure 21 shows exemplary cytosine editing by CBE4-nNme2Cas9 (D16A)-UGI-UGI. Upper panel shows the KANK3 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples. Bottom panel shows the quantification of the substitution rate of each type of base in the CBE4- nNme2Cas9 (D16A)-UGI-UGI editing window of the KANK3 target sequences.
Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red.
Figure 22 shows exemplary cytosine and adenine editing by CBE4-nNme2Cas9 (D16A)- UGI-UGI and optimized ABEmax-nNme2Cas9 (D16A), respectively. Upper panel shows the PLXNB2 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples. Middle panel shows the quantification of the substitution rate of each type of base in the optimized ABEmax-nNme2Cas9 (D16A) editing windows of the PLXNB2 target sequences. Sequence tables show nucleotide frequencies at each position. Frequencies of expected A-to-G conversion are highlighted in red. Bottom panel shows the quantification of the substitution rate of each type of base in the CBE4-nNme2Cas9 (D16A)- UGI-UGI editing windows of the PLXNB2 target sequences. Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red. Detailed Description Of The Invention
The present invention is related to the field of gene editing. In particular, the gene editing is directed toward single nucleotide base editing. For example, such single nucleotide base editing results in a conversion of a C•G base pair to a T•A base pair. The high accuracy and precision of the presently disclosed single nucleotide base gene editor is accomplished by an NmeCas9 nuclease that is fused to a nucleotide deaminase protein. The compact nature of the NmeCas9 coupled with a larger number of compatible protospacer adjacent motifs provide the Cas9 fusion constructs contemplated herein can edit sites that are not targetable by conventional SpyCas9 base editor platforms. A. NmeCas9 Single Base Editing
Cas9 is a programmable nuclease that uses a guide RNA to create a double-stranded break at any desired genomic locus. This programmability has been harnessed for biomedical and therapeutic approaches. However, Cas9-induced breaks often lead to imprecise repair by the cellular machinery, hindering its therapeutic application for single-base corrections as well as uniform and precise gene knock-outs. Moreover, it is extremely challenging to combine Cas9- induced DNA double strand breaks and a repair template for homologous directed repair (HDR) for correcting genetic mutations in post-mitotic cells (e.g. neuronal cells).
Single nucleotide base editing is a genome editing approach where a nuclease-dead or - impaired Cas9 (e.g., dead Cas9 (dCas9) or nickase Cas9 (nCas9)) is fused to another enzyme capable of base-editing nucleotides without causing DNA double strand breaks. To date, two broad classes of Cas9 base editors have been developed: i) cytidine deaminase (edits a C•G base pair to a T•A base pair) SpyCas9 fusion protein; and ii) adenosine deaminase (edits a A•T base pair to a G•C base pair) SpyCas9. Liu et al.,“Nucleobase editors and uses thereof” US
2017/0121693; and Lui et al.,“Fusions of cas9 domains and nucleic acid-editing domains” US 2015/0166980; (both herein incorporated by reference).
However as mentioned above, SpyCas9 base editing platforms cannot be used to target all single-base mutations due to their limited editing windows. The editing window is constrained by the requirement for an NGG PAM. SpyCas9 is also intrinsically associated with high off-targeting effects in genome editing.
In one embodiment, the present invention contemplates a deaminase fusion protein with a compact and hyper-accurate Nme2Cas9 (Neisseria meningitidis spp.). This Nme2Cas9 has 1,082 amino acids as compared to SpyCas9 that has 1,368 amino acids. This Nme2Cas9 ortholog functions efficiently in mammalian cells, recognizes an N4CC PAM, and is intrinsically hyper- accurate. Edraki et al., Mol Cell. (in preparation).
Although it is not necessary to understand the mechanism of an invention, it is believed that the compactness and hyper-accuracy of an NmeCas9 base editor targets single-base mutations that could not be reached previously by other Cas9 platforms currently known in the art. It is further believed that the NmeCas9 base editors contemplated herein target pathogenic mutations that are not feasible via current base editor platforms, and with an increased base editing accuracy. In one embodiment, the present invention contemplates a fusion protein comprising a Nme2Cas9 and a deaminase protein, exemplary examples including ABE7.10-nNme2Cas9 (D16A); Optimized nNme2Cas9-ABEmax; nNme2Cas9-CBE4 (equals BE4-nNme2Cas9 (D16A)-UGI-UGI ) as well as ABEmax-nNme2Cas9 (D16A). See, Figure 1A, Figure 1B, Figure 1C, Figure 1D and Figure 1E.
Figure 1 illustrates exemplary schematic embodiments of an NmeCas9 deaminase fusion protein single base editor and exemplary constructed plasmids of base editors. Figure 1A shows an exemplary YE1-BE3-nNme2Cas9 (D16A)-UGI construct. Figure 1B shows an exemplary ABE7.10 nNme2Cas9 (D16A) construct. Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct. Figure 1C shows an exemplary ABE7.10-nNme2Cas9 (D16A) construct comprising two SV40 NLS sequences. Figure 1D shows an exemplary nNme2Cas9-CBE4 (also called a BE4-nNme2Cas9 (D16A)-UGI-UGI) construct. Figure 1E shows an exemplary optimized nNme2Cas9-ABEmax construct.
In one embodiment, the deaminase protein is Apobec1 (YE1-BE3). It is not intended to limit Apobec1 to one organism. In one embodiment, the Apobec1 is derived from a rat species. Kim et al.,“Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions”. Nature Biotechnology 35 (2017). In one embodiment, the Nme2Cas9 comprises an nNme2Cas9 D16A mutant. In one embodiment, the fusion protein further comprises a uracil glycosylase inhibitor protein (UGI). In one embodiment, the fusion protein comprises a YE1-BE3-nNme2Cas9 (D16A)-UGI construct. In one embodiment, the YE1-BE3-nNme2Cas9 (D16A)-UGI construct has the sequence of:
MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSIWRHTSQNTNK HVEVNFIEKFTTERYFCPNTRCSITWFLSYSPCGECSRAITEFLSRYPHVTLFIYIARLYHH ADPENRQGLRDLISSGVTIQIMTEQESGYCWRNFVNYSPSNEAHWPRYPHLWVRLYVLE LYCIILGLPPCLNILRRKQPQLTFFTIALQSCHYQRLPPHILWATGLKSGSETPGTSESATP ESMAAFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAMAR RLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPL EWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAELALNKFEK ESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKS KLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSS ELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKR YDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPARIHIET AREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCL YSGKEINLVRLNEKGYVEIDAALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYFNGKDNS REWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHILLTGKG KRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNAF DGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLS SRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMV NYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQESGVLLNKK NAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDDSYTFCFSL HKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQKYQVNEL GKEIRPCRLKKRPPVRSGGSTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESDIL VHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSGGSPKKKRKV* YE1-BE3 (underlined); linker (bold), nNme2Cas9 (italics), UGI (bold/underlined), SV40 NLS (plain). In one embodiment, the YE1-BE3-nNme2Cas9 (D16A)-UGI construct has the sequence of:
MSSETGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSIWRHTSQNTNK HVEVNFIEKFTTERYFCPNTRCSITWFLSYSPCGECSRAITEFLSRYPHVTLFIYIARLYHH ADPENRQGLRDLISSGVTIQIMTEQESGYCWRNFVNYSPSNEAHWPRYPHLWVRLYVLE LYCIILGLPPCLNILRRKQPQLTFFTIALQSCHYQRLPPHILWATGLKSGSETPGTSESATP ESMAAFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAMAR RLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPL EWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAELALNKFEK ESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKS KLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSS ELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKR YDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPARIHIET AREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCL YSGKEINLVRLNEKGYVEIDAALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYFNGKDNS REWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHILLTGKG KRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNAF DGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLS SRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMV NYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQESGVLLNKK NAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDDSYTFCFSL HKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQKYQVNEL GKEIRPCRLKKRPPVRSGGSTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESDI LVHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSGGSPKKKRKV* YE1-BE3 (underlined); linker (bold), nNme2Cas9 (italics), UGI (bold/underlined), SV40 NLS (plain). In one embodiment, the present invention contemplates a fusion protein comprising an NmeCas9/ABE7.10 deaminase protein. In one embodiment, the deaminase protein is TadA. In one embodiment, the deaminase protein is TadA 7.10. In one embodiment, the ABE7.10- nNme2Cas9 (D16A) construct has the following sequence:
MSEVEFSHEYWMRHALTLAKRAWDEREVPVGAVLVHNNRVIGEGWNRPIGRHDPTAH AEIMALRQGGLVMQNYRLIDATLYVTLEPCVMCAGAMIHSRIGRVVFGARDAKTGAAG SLMDVLHHPGMNHRVEITEGILADECAALLSDFFRMRRQEIKAQKKAQSSTDSGGSSGG SSGSETPGTSESATPESSGGSSGGSSEVEFSHEYWMRHALTLAKRARDEREVPVGAV LVLNNRVIGEGWNRAIGLHDPTAHAEIMALRQGGLVMQNYRLIDATLYVTFEPCV MCAGAMIHSRIGRVVFGVRNAKTGAAGSLMDVLHYPGMNHRVEITEGILADECAA LLCYFFRMPRQVFNAQKKAQSSTDSGGSSGGSSGSETPGTSESATPESSGGSSGGSMAAF KPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAMARRLARSVR RLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPLEWSAVLL HLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAELALNKFEKESGHIRN QRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDAVQKMLG HCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKSKLTYAQA RKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSSELQDEIG TAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEI YGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPARIHIETAREVGKSF KDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEINLV RLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKA RVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHILLTGKGKRRVFASN GQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNAFDGKTIDK ETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLSSRPEAVH EYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMVNYKNGR EIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQESGVLLNKKNAYTIA DNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDDSYTFCFSLHKYDLI AFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQKYQVNELGKEIRP CRLKKRPPVREDKRPAATKKAGQAKKKK*
TadA (underlined), TadA 7.10 (underlined/bold), linker (bold), nNme2Cas9 (italics), Nucleoplasmin NLS (plain). In one embodiment, an ABE7.10-nNme2Cas9 (D16A) construct has the following amino acid sequence:
MSEVEFSHEYWMRHALTLAKRAWDEREVPVGAVLVHNNRVIGEGWNRPIGRHDPTAH AEIMALRQGGLVMQNYRLIDATLYVTLEPCVMCAGAMIHSRIGRVVFGARDAKTGAAG SLMDVLHHPGMNHRVEITEGILADECAALLSDFFRMRRQEIKAQKKAQSSTDSGGSSGG SSGSETPGTSESATPESSGGSSGGSSEVEFSHEYWMRHALTLAKRARDEREVPVGAV LVLNNRVIGEGWNRAIGLHDPTAHAEIMALRQGGLVMQNYRLIDATLYVTFEPCV MCAGAMIHSRIGRVVFGVRNAKTGAAGSLMDVLHYPGMNHRVEITEGILADECAA LLCYFFRMPRQVFNAQKKAQSSTDSGGSSGGSSGSETPGTSESATPESSGGSSGGSMA AFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAMARRLARS VRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPLEWSAV LLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAELALNKFEKESGHI RNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDAVQKM LGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKSKLTYA QARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSSELQD EIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEAC AEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPARIHIETAREVG KSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEI NLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQE FKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHILLTGKGKRRVF ASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNAFDGKTI DKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLSSRPEA VHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMVNYKN GREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQESGVLLNKKNAYT IADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDDSYTFCFSLHKYD LIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQKYQVNELGKEIR PCRLKKRPPVREDKRPAATKKAGQAKKKK*
TadA (underlined), TadA 7.10 (underlined/bold), linker (bold italics), nNme2Cas9 (italics), Nucleoplasmin NLS (plain). In one embodiment, an ABEmax-nNme2Cas9 (D16A) construct has the following amino acid sequence:
MKRTADGSEFESPKKKRKVSEVEFSHEYWMRHALTLAKRAWDEREVPVGAVLVHNN RVIGEGWNRPIGRHDPTAHAEIMALRQGGLVMQNYRLIDATLYVTLEPCVMCAGAMIH SRIGRVVFGARDAKTGAAGSLMDVLHHPGMNHRVEITEGILADECAALLSDFFRMRRQ EIKAQKKAQSSTDSGGSSGGSSGSETPGTSESATPESSGGSSGGSSEVEFSHEYWMRHA LTLAKRARDEREVPVGAVLVLNNRVIGEGWNRAIGLHDPTAHAEIMALRQGGLV MQNYRLIDATLYVTFEPCVMCAGAMIHSRIGRVVFGVRNAKTGAAGSLMDVLHYP GMNHRVEITEGILADECAALLCYFFRMPRQVFNAQKKAQSSTDSGGSSGGSSGSETP GTSESATPESSGGSSGGSMAAFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVF ERAEVPKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNT PWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQT GDFRTPAELALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKE GIETLLMTQRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERP LTDTERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRA LEKEGLKDKKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQIS LKALRRIVPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVI NGVVRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSK DILKLRLYEQQHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNK GNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRF LCQFVADHILLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQ QKITRFVRYKEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEE ADTPEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVK RVWLTEIKLADLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKA VRVEKTQESGVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDI DCKGYRIDDSYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQF RISTQNLVLIQKYQVNELGKEIRPCRLKKRPPVREDKRPAATKKAGQAKKKKFEPKKKRK
V*
TadA (underlined), TadA* 7.10 (underlined/bold), linker (bold italics), nNme2Cas9 (italics), Nucleoplasmin NLS (plain) and SV40 NLS (BOLD). In one embodiment, a CBE4-nNme2Cas9 (D16A)-UGI-UGI construct has the following amino acid sequence:
PAAKRVKLDGGSGGGSGGGSGPAAKRVKLDGGSGGGSGGGSGPLEPKKKRKVPWSSE TGPVAVDPTLRRRIEPHEFEVFFDPRELRKETCLLYEINWGGRHSIWRHTSQNTNKHVEV NFIEKFTTERYFCPNTRCSITWFLSWSPCGECSRAITEFLSRYPHVTLFIYIARLYHHADPR NRQGLRDLISSGVTIQIMTEQESGYCWRNFVNYSPSNEAHWPRYPHLWVRLYVLELYCII LGLPPCLNILRRKQPQLTFFTIALQSCHYQRLPPHILWATGLKSGGSSGGSSGSETPGTSE SATPESSGGSSGGSIDKLAAFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFER AEVPKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPW QLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGD FRTPAELALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIE TLLMTQRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTD TERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEK EGLKDKKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKA LRRIVPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGV VRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILK LRLYEQQHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQ TPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQ FVADHILLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKI TRFVRYKEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADT PEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVW LTEIKLADLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRV EKTQESGVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCK GYRIDDSYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRIST QNLVLIQKYQVNELGKEIRPCRLKKRPPVRVYPYDVPDYAGYPYDVPDYAGSYPYDVPDYA GSAAPAAKKKKLDFESGEFLQPGIDLSQLGGDSGGSGGSGGSTNLSDIIEKETGKQLVI QESILMLPEEVEEVIGNKPESDILVHTAYDESTDENVMLLTSDAPEYKPWALVIQDS NGENKIKMLSGGSGGSGGSTNLSDIIEKETGKQLVIQESILMLPEEVEEVIGNKPESD ILVHTAYDESTDENVMLLTSDAPEYKPWALVIQDSNGENKIKMLSGGSPKKKRKVSR GSAAPAAKRVKLDGGSGGGSGGGSGSGPAAKRVKLD
rApobec1 (underlined), UGI (underlined/bold), linker (bold italics), nNme2Cas9 (D16A) (italics), Cmyc-NLS (plain) and SV40 NLS (BOLD). In one embodiment, an optimized nNme2Cas9-ABEmax construct refers to an optimized version with improved promoter, NLS sequences, and linker sequences. In some embodiments, an optimized nNme2Cas9-ABEmax construct comprises, 5' to 3', a C-myc NLS, 12aa linker, 15aa linker, SV40 NLS, TadA, TadA*7.10, 48aa linker, nNme2Cas9, a 73aa linker (3xHA-tag), 15aa linker, and a C-myc NLS. In some embodiments, an optimized nNme2Cas9-ABEmax construct further comprises at least two each alternating C-myc NLS and a 12aa linker at the 3' end. In some embodiments, an optimized nNme2Cas9-ABEmax construct further comprises at least two each alternating 15aa linker and C-myc NLS at the 5' end. See, Figure 1E for example.
In one embodiment, an optimized nNme2Cas9-ABEmax construct has the following amino acid sequence:
PAAKRVKLDGGSGGGSGGGSGPAAKRVKLDGGSGGGSGGGSGPLEPKKKRKV SEVEFSHEYWMRHALTLAKRAWDEREVPVGAVLVHNNRVIGEGWNRPIGRHDPTAHA EIMALRQGGLVMQNYRLIDATLYVTLEPCVMCAGAMIHSRIGRVVFGARDAKTGAAGS LMDVLHHPGMNHRVEITEGILADECAALLSDFFRMRRQEIKAQKKAQSSTDSGGSSGGS SGSETPGTSESATPESSGGSSGGSSEVEFSHEYWMRHALTLAKRARDEREVPVGAVL VLNNRVIGEGWNRAIGLHDPTAHAEIMALRQGGLVMQNYRLIDATLYVTFEPCVM CAGAMIHSRIGRVVFGVRNAKTGAAGSLMDVLHYPGMNHRVEITEGILADECAAL LCYFFRMPRQVFNAQKKAQSSTDSGGSSGGSSGSETPGTSESATPESSGGSSGGSMAA FKPNPINYDIDKLAAFKPNPINYILGLAIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPK TGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAA ALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPA ELALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMT QRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERAT LMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLK DKKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRI VPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRY GSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLY EQQHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYE YFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVA DHILLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRF VRYKEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEK LRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEI KLADLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKT QESGVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRI DDSYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLV LIQKYQVNELGKEIRPCRLKKRPPVRVYPYDVPDYAGYPYDVPDYAGSYPYDVPDYAGSAA PAAKKKKLDFESGEFLQPGGSTSSRGSAAPAAKRVKLDGGSGGGSGGGSGSGPAAKRV KLD
hTadA7.10 (underlined), hTadA*7.10 (underlined/bold), linker (bold italics),
nNme2Cas9 (italics), Cmyc-NLS (plain), SV40-NLS (bold). In some embodiments, a plasmid nSpCas9-ABEmax (Addgene ID:112095) was used for experimental controls and for molecular cloning. In some embodiments, a plasmid nSpCas9- CBE4 (Addgene ID: 100802) was used for experimental controls and for molecular cloning. Electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3- Nme2Cas9 nucleotide deaminase fusion protein achieved robust single-base editing of a C•G base pair to a T•A base pair at an endogenous target site (TS25). See, Figures 2A-C.
Figure 2 presents exemplary data of the electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3-nNme2Cas9 (D16A)-UGI fusion protein efficiently converting C to T at endogenous target site 25 (TS25) in HEK293T cells via nucleofection. Figure 2A shows exemplary sequences for a TS25 endogenous target site (within the black rectangle). GN23 sgRNA base-pairs with the target DNA strand, leaving the displaced DNA strand for cytidine deaminase to edit (e.g. new green nucleotides). Figure 2B shows exemplary sequencing data showing a doublet nucleotide peak (7th position from 5’ end; arrow) demonstrating the successful single base editing of a cytidine to a thymidine (e.g., a C•G base pair conversion to a T•A base pair). Figure 2C shows an exemplary quantitation of the data shown in Figure 2B plotting the percent conversion of C ® T single base editing. The percentage of C converted to T is about 40% in the base editor- and sgRNA-treated sample (p-value = 6.88 x 10-6). The“no sgRNA” control displays the background noise due to Sanger sequencing. EditR (Kluesner et al., 2018) was used to perform the analysis.
Four other YE1-BE3-nNme2Cas9/D16A mutant fusion proteins were co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line expressing enhanced green fluorescent protein (EGFP). Each YE1-BE3-nNme2Cas9/D16A mutant fusion protein had a specific UGI target site. See, Figures 3A-D.
Deep-sequencing analysis indicates YE1-BE3-nNme2Cas9 converts C residues to T residues at each of the four EGFP target sites. The percentage of editing ranged from 0.24% to 2%. The potential base editing window is from nucleotides 2-8 in the displaced DNA strand, counting the nucleotide at the 5’ (PAM-distal) end as nucleotide #1. See, Figures 3A-D.
Figure 3 presents exemplary specific UGI target sites that were respectively integrated into YE1-BE3-nNme2Cas9/D16A mutant fusion proteins and co-expressed with enhanced green fluorescent protein (EGFP) in a stable K562-derived cell line. Converted bases are highlighted in orange color. Background signals were filtered using negative control samples (YE1-BE3- nNme2Cas9 nucleofected K562 cells without sgRNA constructs). N4CC PAMs are boxed. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. Figure 3A shows an exemplary EGFP-Site 1. Figure 3B shows an exemplary EGFP-Site 2. Figure 3C shows an exemplary EGFP-Site 3. Figure 3D shows an exemplary EGFP-Site 4.
Electroporation of HEK293T cells with DNA plasmids comprising a YE1-BE3- nNme2Cas9 c-fos promoter achieved robust single-base editing of a CxG base pair to a TxA base pair at endogenous target sites in the c-fos promoter (Figure 3E). Figure 3E shows an exemplary deep-sequencing analysis indicating where YE1-BE3-nNme2Cas9 converts C residues to T residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange or yellow color. Background signals were filtered using negative control samples. The highest percentage of editing is 32.50%. Figure 3F shows an exemplary deep-sequencing analysis indicating where ABE7.10-nNme2Cas9 or ABEmax (Koblan et al., 2018)-nNme2Cas9 converts A residues to G residues at endogenous c-fos promoter region. The percentage of total reads exhibiting mutations in base-editor-targeted sites is shown in the right column. The converted bases are highlighted in orange color. Background signals were filtered using negative control samples. The percentage of editing is 0.53% by ABE7.10-nNme2Cas9 or 2.33% by ABEmax-nNme2Cas9 (D16A).
In one embodiment, the present invention contemplates the expression of an ABE7.10- nNme2Cas9 (D16A) fusion protein for base editing. Although it is not necessary to understand the mechanism of an invention, it is believed that Nme2Cas9 base editing may be an effective treatment for tyrosinemia by reversing a G-to-A point mutation in the Fah gene with an
ABE7.10-nNme2Cas9 (D16A) fusion protein.
G-to-A mutation (red) at the last nucleotide of exon 8 in Fah gene, causing exon skipping. FAH deficiency leads to toxin accumulation and severe liver damage. The position of a SpyCas9 PAM (black rectangular box) downstream of the mutation is not optimal for designing the sgRNA since the A mutation is out of the efficient base editing window of ABE7.10, which is 4-7th nt at the 5’ (PAM-distal) end (underlined) (Gaudelli et al., 2017).
However, there are two Nme2Cas9 PAMs (red rectangular box) in the downstream sequences that can potentially correct the mutation and revert DNA sequence to wildtype via ABE7.10-nNme2Cas9 (D16A). See, Figure 4.
Figure 4 presents an exemplary alignment of the wildtype Fah gene with the tyrosinemia Fah mutant gene showing an A-G single base gene editing target site (position 9). The respective SpyCas9 single PAM site and NmeCas9 double PAM sites are indicated for demonstrating the suboptimal targeting window relative to the SpyCas9 PAM site. This figure serves as a potential example of a site where Nme2Cas9 could overcome limitations of existing base editors. It is further believed that the NmeCas9 base editor described herein can perform precise base editing that cannot be achieved with conventional SpyCas9-derived base editors due to a suboptimal base editing window relative to available PAMs nearby.
Furthermore, we contemplate extending base editing to a tyrosinemia mouse model for reversing the G-to-A point mutation by viral delivery methods using ABEmax-nNme2Cas9 (D16A), where the desired editing cannot be achieved with SpyCas9-derived base editors due to a suboptimal base editing window relative to available PAMs nearby (e.g. Figure 4).
B. NmeCas9 Constructs: Compact & Hyperaccurate
Clustered, regularly interspaced, short, palindromic repeats (CRISPR) along with CRISPR-associated (Cas) proteins constitute bacterial and archaeal adaptive immune pathways against phages and other mobile genetic elements (MGEs) (Barrangou et al., 2007; Brouns et al., 2008; Marraffini and Sontheimer, 2008). In Type II CRISPR systems, CRISPR RNA (crRNA) is bound to a trans-activating crRNA (tracrRNA) and loaded onto a Cas9 effector protein that cleaves MGE nucleic acids complementary to the crRNA (Garneau et al., 2010; Deltcheva et al., 2011; Sapranauskas et al., 2011; Gasiunas et al., 2012; Jinek et al., 2012). The crRNA:tracrRNA hybrid can be fused into a single-guide RNA (sgRNA) (Jinek et al., 2012). The RNA
programmability of Cas9 endonucleases has made it a powerful genome editing platform in biotechnology and medicine (Cho et al., 2013; Cong et al., 2013; Hwang et al., 2013; Jiang et al., 2013; Jinek et al., 2013; Mali et al., 2013b).
In addition to sgRNA, Cas9 target recognition is usually associated with a 1-5 nucleotide signature downstream of the complementary DNA sequence, called a protospacer adjacent motif (PAM) (Deveau et al., 2008; Mojica et al., 2009). Cas9 orthologs exhibit considerable diversity in PAM length and sequence. Among Cas9 orthologs that have been characterized,
Streptococcus pyogenes Cas9 (SpyCas9) is the most widely used, in part because it recognizes a short NGG PAM (Jinek et al., 2012) (N represents any nucleotide) that affords a high density of targetable sites. Nevertheless, Spy’s relatively large size (i.e., 1,368 amino acids) makes this Cas9 difficult to package (along with sgRNA and promoters) into a single recombinant adeno- associated virus (rAAV). This has been shown to be a drawback for therapeutic applications given the promise shown by AAV vectors for in vivo gene delivery (Keeler et al., 2017).
Moreover, SpyCas9 and its RNA guides have required extensive characterization and engineering to minimize the tendency to edit near-cognate, off-target sites. (Bolukbasi et al., 2015b; Tsai and Joung, 2016; Tycko et al., 2016; Chen et al., 2017; Casini et al., 2018; Yin et al., 2018). To date, subsequent engineering efforts have not overcome these size limitations.
Several Cas9 orthologs of less than 1,100 amino acids in length obtained from diverse species have been validated for mammalian genome editing, including strains of N. meningitidis (NmeCas9, 1,082 aa) (Esvelt et al., 2013; Hou et al., 2013), Staphylococcus aureus (SauCas9, 1,053 aa) (Ran et al., 2015), Campylobacter jejuni (CjeCas9, 984 aa) (Kim et al., 2017), and Geobacillus stearothermophilus (GeoCas9, 1,089 aa) (Harrington et al., 2017b). NmeCas9, CjeCas9, and GeoCas9 are representatives of type II-C Cas9s (Mir et al., 2018), most of which are <1,100 aa. With the exception of GeoCas9, each of these shorter sequence orthologs has been successfully deployed for in vivo editing via all-in-one AAV delivery (in which a single vector expresses both guide and effector) (Ran et al., 2015; Kim et al., 2017; Ibraheim et al., 2018, submitted). Furthermore, NmeCas9 and CjeCas9 have been shown to be naturally resistant to off-target editing (Lee et al., 2016; Kim et al., 2017; Amrani et al., 2018, submitted).
However, the PAMs that are recognized by compact Cas9s are usually longer than that of SpyCas9, substantially reducing the number of targetable sites at or near a given locus; for example, i) N4GAYW/N4GYTT/N4GTCT for NmeCas9 (Esvelt et al., 2013; Hou et al., 2013; Lee et al., 2016; Amrani et al., 2018); ii) N2GRRT for SauCas9 (Ran et al., 2015); iii) N4RYAC for CjeCas9 (Kim et al., 2017); and iv) N4CRAA/N4GMAA for GeoCas9s (Harrington et al., 2017b) (Y = C, T; R = A, G; M = A, C; W = A, T). A smaller subset of target sites is
advantageous for highly accurate and precise gene editing tasks including, but not limited to: i) editing of small targets (e.g. miRNAs); ii) correction of mutations by base editing which alters a very narrow window of bases relative to the PAM (Komor et al., 2016; Gaudelli et al., 2017); or iii) precise editing via homology-directed repair (HDR) which is most efficient when the rewritten bases are close to the cleavage site (Gallagher and Haber, 2018). Because of PAM restrictions, many editing sites cannot be targeted using all-in-one AAV vectors for in vivo delivery even with these shorter Cas9 proteins. For example, A SauCas9 mutant (SauCas9KKH) has been developed that has reduced PAM constraints (N3RRT), though this increase in targeting range often comes at the cost of reduced on-target editing efficacy, and off-target edits are still observed. (Kleinstiver et al., 2015).
Safe and effective CRISPR-based therapeutic gene editing will be greatly enhanced by Cas9 orthologs and variants that are highly active in human cells, resistant to off-targeting, sufficiently compact for all-in-one AAV delivery, and capable of accessing a high density of genomic sites. In one embodiment, the present invention contemplates a compact, hyper-accurate Cas9 (Nme2Cas9) from a distinct strain of N. meningitidis. In one embodiment, the present invention contemplates a method for single-AAV delivery of Nme2Cas9 and its sgRNA to perform efficient genome editing in vivo and/or ex vivo. Although it is not necessary to understand the mechanism of an invention, it is believed that this ortholog functions efficiently in mammalian cells and recognizes an N4CC PAM that affords a target site density identical to that of wild-type SpyCas9 (e.g., every 8 bp on average, when both DNA strands are considered).
1. PAM Interacting Domains And Anti-CRISPR Proteins PAM recognition by Cas9 orthologs occurs predominantly through protein-DNA interactions between the PAM Interacting Domain (PID) and the nucleotides adjacent to the protospacer (Jiang and Doudna, 2017). PAM mutations often enable phage escape from type II CRISPR immunity (Paez-Espino et al., 2015), placing these systems under selective pressure not only to acquire new CRISPR spacers, but also to evolve new PAM specificities via PID mutations. In addition, some phages and MGEs express anti-CRISPR (Acr) proteins that inhibit Cas9 (Pawluk et al., 2016; Hynes et al., 2017; Rauch et al., 2017). PID binding is an effective inhibitory mechanism adopted by some Acrs (Dong et al., 2017; Shin et al., 2017; Yang and Patel, 2017), suggesting that PID variation may also be driven by selective pressure to escape Acr inhibition. Cas9 PIDs can evolve such that closely-related orthologs recognize distinct PAMs, as illustrated recently in two species of Geobacillus. The Cas9 encoded by G.
stearothermophilus recognizes a N4CRAA PAM, but when its PID was swapped with that of strain LC300’s Cas9, its PAM requirement changed to N4GMAA (Harrington et al., 2017b).
In one embodiment, the present invention contemplates a plurality of N. meninigitidis Cas9 orthologs with divergent PIDs that recognize different PAMs. In one embodiment, the present invention contemplates a Cas9 protein with a high sequence identity (>80% along their entire lengths) to that of NmeCas9 strain 8013 (Nme1Cas9) (Zhang et al., 2013). Nme1Cas9 also has a small size and naturally high accuracy as discussed above. (Lee et al., 2016; Amrani et al., 2018). Alignments revealed three clades of meningococcal Cas9 orthologs, each with >98% identity in the N-terminal ~820 amino acid (aa) residues, which includes all regions of the protein other than the PID. See, Figure 5A and Figure 6A.
All of these Cas9 orthologs are 1,078-1,082 aa in length. The first clade (group 1) includes orthologs in which the >98% aa sequence identity with Nme1Cas9 extends through the PID. In contrast, the other two groups had PIDs that were significantly diverged from that of Nme1Cas9, with group 2 and group 3 orthologs averaging ~52% and ~86% PID sequence identity with Nme1Cas9, respectively. One meningococcal strain was selected from each group: i) De11444 from group 2; and ii) 98002 from group 3 for detailed analysis, which are referred to herein as Nme2Cas9 (1,082 aa) and Nme3Cas9 (1,081 aa), respectively. The CRISPR-cas loci from these two strains have repeat sequences and spacer lengths that are identical to those of strain 8013. See, Figure 6B. This strongly suggested that their mature crRNAs also have 24nt guide sequences and 24 nt repeat sequences (Zhang et al., 2013). Similarly, the tracrRNA sequences of De11444 and 98002 were 100% identical to the 8013 tracrRNA. See, Figure 6B. These observations imply that the same sgRNA sequence scaffold can guide DNA cleavage by all three Cas9s.
To determine whether these Cas9 orthologs have distinct PAMs, the PID of Nme1Cas9 was replaced with that of either Nme2Cas9 or Nme3Cas9. To identify the corresponding PAM requirements, these protein chimeras were expressed in Escherichia coli, purified, and used for in vitro PAM identification (Karvelis et al., 2015; Ran et al., 2015; Kim et al., 2017). Briefly, a pool of DNA fragments containing a protospacer followed by a 10-nt randomized sequence was cleaved in vitro using recombinant Cas9 and a cognate, in vitro-transcribed sgRNA. See, Figure 5B. Only those DNAs containing a Cas9 PAM sequence were expected to be cleaved. Cleavage products were then sequenced to identify the PAMs. See, Figures 5C-D.
The expected N4GATT PAM consensus was validated in the recovered full-length Nme1Cas9. See, Figure 5C. Chimeric PID-swapped derivatives exhibited a strong preference for a C residue in the 5th position in place of the G recognized by Nme1Cas9. See, Figure 5D.
In one embodiment, ABE7.10-nNme2Cas9 (D16A) is used for single-base editing of AxT base pair to a GxC base pair. In one embodiment, BEmax-nNme2Cas9 (D16A) is used for single-base editing of AxT base pair to a GxC base pair. (See, Figure 3F). Figure 5 illustrates exemplary three closely related Neisseria meningitidis Cas9 orthologs that have distinct PAMs. Figure 5A shows an exemplary schematic showing mutated residues (orange spheres) between Nme2Cas9 (left) and Nme3Cas9 (right) mapped onto the predicted structure of Nme1Cas9, revealing the cluster of mutations in the PID (black). Figure 5B shows an exemplary experimental workflow of the in vitro PAM discovery assay with a 10-bp randomized PAM region. Following in vitro digestion, adapters were ligated to cleaved products for library construction and sequencing. Figure 5C shows exemplary sequence logos resulting from in vitro PAM discovery reveal the enrichment of a N4GATT PAM for Nme1Cas9, consistent with its previously established specificity. Figure 5D shows exemplary sequence logos indicating that Nme1Cas9 with its PID swapped with that of Nme2Cas9 (left) or Nme3Cas9 (right) requires a C at PAM position 5. The remaining nucleotides were not determined with high confidence due to the modest cleavage efficiency of the PID-swapped protein chimeras (see Figure 6C). Figure 5E shows an exemplary sequence logo showing that full-length Nme2Cas9 recognizes an N4CC PAM, based on efficient substrate cleavage of a target pool with a fixed C at PAM position 5, and with PAM nts 1-4 and 6-8 randomized.
Any remaining PAM nucleotides could not be confidently assigned due to the low cleavage efficiencies of the chimeric proteins under the conditions used. See, Figure 6C. To further resolve the PAMs, in vitro assays were performed on a library with a 7-nt randomized sequence possessing an invariant C at the 5th PAM position (e.g., 5’-NNNNCNNN-3’ on the sgRNA-noncomplementary strand). This strategy yielded a much higher cleavage efficiency and the results indicated that the Nme2Cas9 and Nme3Cas9 PIDs recognize NNNNCC(A) and NNNNCAAA PAMs, respectively. See, Figures 6C-D. The Nme3Cas9 consensus is similar to that of GeoCas9 (Harrington et al., 2017b).
These tests were repeated using a full-length Nme2Cas9 (rather than a PID-swapped chimera) with the NNNNCNNN DNA pool, and again a NNNNCC(A) consensus was recovered. See, Figure 5E. It was noted that this test had more efficient cleavage. See, Figure 6C. These data suggest that one or more of the 15 amino acid changes in Nme2Cas9 (relative to Nme1Cas9) outside of the PID support efficient DNA cleavage activity. See, Figure 6C. Because the unique, 2-3 nt PAM of Nme2Cas9 affords a higher density of potential target sites than the previously described compact Cas9 orthologs, it was selected for further analyses. Figure 6 presents a characterization of Neisseria meningitidis Cas9 orthologs with rapidly-evolving PIDs, as related to Figure 5. Figure 6A shows an exemplary unrooted phylogenetic tree of NmeCas9 orthologs that are >80% identical to Nme1Cas9. Three distinct branches emerged, with the majority of mutations clustered in the PID. Groups 1 (blue), 2 (orange), and 3 (green) have PIDs with >98%, approximately 52%, and approximately86% identity to Nme1Cas9, respectively. Three representative Cas9 orthologs (one from each group) (Nme1Cas9, Nme2Cas9 and Nme3Cas9) are indicated. Figure 6B shows an exemplary schematic showing the CRISPR-cas loci of the strains encoding the three Cas9 orthologs (Nme1Cas9, Nme2Cas9, and Nme3Cas9) from (A). Percent identities of each CRISPR-Cas component with N. meningitidis 8013 (encoding Nme1Cas9) are shown. Blue and red arrows denote pre-crRNA and tracrRNA transcription initiation sites, respectively. Figure 6C shows an exemplary normalized read counts (% of total reads) from cleaved DNAs from the in vitro assays for intact Nme1Cas9 (grey), for chimeras with Nme1Cas9’s PID swapped with those of
Nme2Cas9 and Nme3Cas9 (mixed colors), and for full-length Nme2Cas9 (orange), are plotted. The reduced normalized read counts indicate lower cleavage efficiencies in the chimeras. Figure 6D shows an exemplary sequence logos from the in vitro PAM discovery assay on an
NNNNCNNN PAM pool by Nme1Cas9 with its PID swapped with those of Nme2Cas9 (left) or Nme3Cas9 (right).
2. N4CC PAM-Directed Gene Editing
To test the efficacy of Nme2Cas9 in human genome editing, a full-length (e.g., not PID- swapped) human-codon-optimized Nme2Cas9 construct was cloned into a mammalian expression plasmid with appended nuclear localization signals (NLSs) and linkers validated previously for Nme1Cas9 (Amrani et al., 2018). For initial tests, a modified, fluorescence-based Traffic Light Reporter (TLR2.0) was used (Certo et al., 2011). Briefly, a disrupted GFP is followed by an out-of-frame T2A peptide and mCherry cassette. When DNA double-strand breaks (DSBs) are introduced in the broken-GFP cassette, a subset of non-homologous end joining (NHEJ) repair events leave +1-frameshifted indels, placing mCherry in frame and yielding red fluorescence that can be easily quantified by flow cytometry See, Figure 7A.
Homology-directed repair (HDR) outcomes can also be scored simultaneously by including a DNA donor that restores the functional GFP sequence, yielding a green fluorescence (Certo et al., 2011). Because some indels do not introduce a +1 frameshift, the fluorescence readout generally provides an underestimate of the true editing efficiency. Nonetheless, the speed, simplicity, and low cost of the assay makes it useful as an initial, semi-quantitative measure of genome editing in HEK293T cells carrying a single TLR2.0 locus incorporated via lentivector.
For initial tests, Nme2Cas9 plasmid was transiently co-transfected with one of fifteen sgRNA plasmids carrying spacers that target TLR2.0 sites with N4CC PAMs. No HDR donor was included, so only NHEJ-based editing (mCherry) was scored. Most sgRNAs were in a G23 format (i.e. a 5’-terminal G to facilitate transcription, followed by a 23nt guide sequence), as used routinely for Nme1Cas9 (Lee et al., 2016; Pawluk et al., 2016; Amrani et al., 2018;
Ibraheim et al., 2018). No sgRNA and an sgRNA targeting an N4GATT PAM were used as negative controls, and SpyCas9+sgRNA and Nme1Cas9+sgRNA co-transfections (targeting NGG and N4GATT protospacers, respectively) were included as positive controls. Editing by SpyCas9 and Nme1Cas9 was readily detectable (~28% and 10% mCherry, respectively). See, Figure 7B.
For Nme2Cas9, all 15 targets with N4CC PAMs were functional, though to various extents ranging from 4% to 20% mCherry. These fifteen sites include examples with each of the four possible nucleotides in the 7th PAM position (e.g., after the CC dinucleotide), indicating that a slight preference for an A residue was observed in vitro (Figure 5E) does not reflect a PAM requirement for editing applications in human cells. The N4GATT PAM control yielded mCherry signal similar to no-sgRNA control. See, Figure 7B.
To determine whether both C residues in the N4CC PAM are involved in editing, a series of N4DC (D = A, T, G) and N4CD PAM sites were tested in TLR2.0 reporter cells. See, Figures 8A and 8B. No detectable editing was found at any of these sites, providing an initial indication that both C residues of the N4CC PAM consensus are required for efficient Nme2Cas9 activity.
The length of the spacer in the crRNA differs among Cas9 orthologs and can affect on- vs. off-target activity (Cho et al., 2014; Fu et al., 2014). SpyCas9’s optimal spacer length is 20 nts, with truncations down to 17 nts tolerated (Fu et al., 2014). In contrast, Nme1Cas9 usually has 24-nt spacers (Hou et al., 2013; Zhang et al., 2013), and tolerates truncations down to 18-20 nts (Lee et al., 2016; Amrani et al., 2018). To test spacer length requirements for Nme2Cas9, guide RNA plasmids were created for each targeted single TLR2.0 site, but with varying spacer lengths. See, Figure 7C and Figure 8C. Comparable activities were observed with G23, G22 and G21 guides, but significantly decreased activity upon further truncation to G20 and G19 lengths. See, Figure 7C. These results validate Nme2Cas9 as a genome editing platform, with 22-24 nt guide sequences, at N4CC PAM sites in cultured human cells.
Figure 7 presents exemplary data showing that Nme2Cas9 uses a 22-24 nt spacer to edit sites adjacent to an N4CC PAM. All experiments were done in triplicate, and error bars represent the standard error of the mean (s.e.m.). Figure 7A shows an exemplary schematic diagram depicting transient transfection and editing of HEK293T TLR2.0 cells, with mCherry+ cells detected by flow cytometry 72 hours after transfection. Figure 7B shows an exemplary
Nme2Cas9 editing of the TLR2.0 reporter. Sites with N4CC PAMs were targeted with varying efficiencies, while no Nme2Cas9 targeting was observed at an N4GATT PAM or in the absence of sgRNA. SpyCas9 (targeting a previously validated site with an NGG PAM) and Nme1Cas9 (targeting N4GATT) were used as positive controls. Figure 7C shows an exemplary effect of spacer length on the efficiency of Nme2Cas9 editing. An sgRNA targeting a single TLR2.0 site, with spacer lengths varying from 24 to 20 nts (including the 5’-terminal G required by the U6 promoter), indicate that highest editing efficiencies are obtained with 22-24 nt spacers. Figure 7D shows an exemplary An Nme2Cas9 dual nickase can be used in tandem to generate NHEJ- and HDR-based edits in TLR2.0. Nme2Cas9- and sgRNA-expressing plasmids, along with an 800-bp dsDNA donor for homologous repair, were electroporated into HEK293T TLR2.0 cells, and both NHEJ (mCherry+) and HDR (GFP+) outcomes were scored by flow cytometry. HNH nickase, Nme2Cas9D16A; RuvC nickase, Nme2Cas9H588A. Cleavage sites 32 bp and 64 bp apart were targeted using either nickase. The HNH nickase (Nme2Cas9D16A) yielded efficient editing, particularly with the cleavage sites that were separated by 32 bp, whereas the RuvC nickase (Nme2Cas9H588A) was not effective. Wildtype Nme2Cas9 was used as a control.
3. Precise Editing By HDR And HNH Nickase
Cas9 enzymes use their HNH and RuvC domains to cleave the guide-complementary and non-complementary strand of the target DNA, respectively. SpyCas9 nickases (nCas9s), in which either the HNH or RuvC domain is mutationally inactivated, have been used to induce homology-directed repair (HDR) and to improve genome editing specificity via DSB induction by dual nickases (Mali et al., 2013a; Ran et al., 2013).
To test the efficacy of Nme2Cas9 as a nickase, a Nme2Cas9D16A (HNH nickase) and Nme2Cas9H588A (RuvC nickase) were created, which possess alanine mutations in catalytic residues of the RuvC and HNH domains, respectively (Esvelt et al., 2013; Hou et al., 2013; Zhang et al., 2013). TLR2.0 cells, along with a GFP donor dsDNA, were used to determine whether Nme2Cas9-induced nicks can induce precise edits via HDR. Target sites within TLR2.0 were used to test the functionality of each nickase using guides targeting cleavage sites spaced 32 bp and 64 bp apart. See, Figure 7D. Wildtype Nme2Cas9 targeting a single site showed efficient editing, with both NHEJ and HDR as outcomes of repair. For nickases, cleavage sites 32 bp and 64 bp apart showed editing using the Nme2Cas9D16A (HNH nickase), but neither target pair worked with Nme2Cas9H588A. These results suggest that Nme2Cas9 HNH nickase can be used for efficient genome editing, as long as the sites are in close proximity.
Studies in previously characterized Cas9s have identified a specific region proximal to the PAM where Cas9 activity is highly sensitive to sequence mismatches. This 8 to 12-nt region is known as the seed sequence and has been observed among all Cas9s characterized to date (Gorski et al., 2017). To determine whether Nme2Cas9 also possesses a seed sequence, a series of transient transfections was performed, each targeting the same locus in TLR2.0, but with a single-nucleotide mismatch at different positions of the guide. See, Figure 8D. A significant decrease in the number of mCherry-positive cells was observed for mismatches in the first 10-12 nts proximal to the PAM, suggesting that Nme2Cas9 possesses a seed sequence in this region.
Figure 8 presents exemplary data showing PAM, spacer, and seed requirements for Nme2Cas9 targeting in mammalian cells, as related to Figure 7. All experiments were done in triplicate and error bars represent s.e.m. Figure 8A shows an exemplary Nme2Cas9 targeting at N4CD sites in TLR2.0, with editing estimated based on mCherry+ cells. Four sites for each non- C nucleotide at the tested position (N4CA, N4CT and N4CG) were examined, and an N4CC site was used as a positive control. Figure 8B shows an exemplary Nme2Cas9 targeting at N4DC sites in TLR2.0 [similar to (A)]. Figure 8C shows exemplary guide truncations on a TLR2.0 site (distinct from that in Figure 2C) with a N4CCA PAM, revealing similar length requirements as those observed at the other site. Figure 8D shows exemplary Nme2Cas9 targeting efficiency is differentially sensitive to single-nucleotide mismatches in the seed region of the sgRNA. Data show the effects of walking single-nucleotide sgRNA mismatches along the 23-nt spacer in a TLR2.0 target site.
4. Delivery Methods To Mammalian Cell Types
Nme2Cas9’s ability to function in different mammalian cell lines was tested using various delivery methods. As an initial test, forty (40) different sites (29 with a N4CC PAM, and 11 sites were tested with a N4CD PAM). Several loci were selected (AAVS1, VEGFA, etc.), and target sites with N4CC PAMs were randomly chosen for editing with Nme2Cas9. Editing (%) was determined by transiently transfecting 150 ng of Nme2Cas9 along with 150 ng of sgRNA plasmids followed by TIDE analysis 72 hours post-transfection. A subset of sites exhibiting a range of editing efficiencies in this initial screen was selected for repeat analyses in triplicate. See, Figure 9A; and Table 1.
Figure 9 presents exemplary data showing Nme2Cas9 genome editing at endogenous loci in mammalian cells via multiple delivery methods. All results represent 3 independent biological replicates, and error bars represent s.e.m. Figure 9A shows an exemplary Nme2Cas9 genome editing of endogenous human sites in HEK293T cells following transient transfection of Nme2Cas9- and sgRNA-expressing plasmids.40 sites were screened initially (Table 1); the 14 sites shown (selected to include representatives of varying editing efficiencies, as measured by TIDE) were then re-analyzed in triplicate. An Nme1Cas9 target site (with an N4GATT PAM) was used as a negative control. Figure 9B shows exemplary data charts: Left panel: Transient transfection of a single plasmid expressing both Nme2Cas9 and sgRNA (targeting the Pcsk9 and Rosa26 loci) enables editing in Hepa1-6 mouse cells, as detected by TIDE. Right panel:
Electroporation of sgRNA plasmids into K562 cells stably expressing Nme2Cas9 from a lentivector results in efficient indel formation. Figure 9C shows exemplary Nme2Cas9 can be electroporated as an RNP complex to induce genome editing.40 picomoles Cas9 along with 50 picomoles of in vitro-transcribed sgRNAs targeting three different loci were electroporated into HEK293T cells. Indels were measured after 72h using TIDE. Table 1. Exemplary Endogenous human genome editing sites targeted by Nme2Cas9.
Figure imgf000051_0001
HEK293T cells were used to support transient transfections and at 72-hours post transfection the, cells were harvested, followed by genomic DNA extraction and selective amplification of the targeted locus. TIDE analysis was used to measure indel efficiency at each locus (Brinkman et al., 2014). Nme2Cas9 editing was detectable at most of these sites, even though efficiencies varied depending on the target sequence. Table 1. Interestingly, Nme2Cas9 induced indels at several genomic sites with N4CD PAMs, albeit less consistently and at lower levels. Table 1. Fourteen (14) sites with N4CC PAMs were analyzed in triplicate, and consistent editing was observed. See, Figure 9A. In addition, editing efficiency could be improved significantly by increasing the quantity of the Nme2Cas9 plasmid delivered, and this high efficiency could be extended to precise segmental deletion with two guides. See, Figures 10A and 10B.
The ability of Nme2Cas9 to function was tested in mouse Hepa1-6 cells (hepatoma- derived). For Hepa1-6 cells, a single plasmid encoding both Nme2Cas9 and an sgRNA (targeting either Rosa26 or Pcsk9) was transiently transfected and indels were measured after 72 hrs.
Editing was readily observed at both sites. See, Figure 9B, left. Nme2Cas9’s functionality was also tested when stably expressed in human leukemia K562 cells. To this end, a lentiviral construct was created expressing Nme2Cas9 and transduced cells to stably express Nme2Cas9 under the control of the SFFV promoter. This stable cell line did not show any visible differences with respect to growth and morphology in comparison to untransduced cells, suggesting that Nme2Cas9 is not toxic when stably expressed. These cells were transiently electroporated with plasmids expressing sgRNAs and analyzed by TIDE after 72 hours to measure indel efficiencies. Efficient (>50%) editing was observed at all three sites tested, validating Nme2Cas9’s ability to function upon lentiviral delivery in K562 cells. See, Figure 9B.
Ribonucleoprotein (RNP) delivery of Cas9 and its sgRNA is also useful for some genome editing applications, and the greater transience of Cas9’s presence can minimize off-target editing (Kim et al., 2014; Zuris et al., 2015). Moreover, some cell types (e.g. certain immune cells) are recalcitrant to DNA transfection-based editing (Schumann et al., 2015). To test whether Nme2Cas9 is functional by RNP delivery, a 6xHis-tagged Nme2Cas9 (fused to three NLSs) was cloned into a bacterial expression construct and the recombinant protein was purified. The recombinant protein was then loaded with T7 RNA polymerase-transcribed sgRNAs targeting three previously validated sites. Electroporation of the Nme2Cas9:sgRNA complex induced successful editing at each of the three target sites in HEK293T cells, as detected by TIDE. See, Figure 9C. Collectively these results indicate that Nme2Cas9 can be delivered effectively via plasmid or lentivirus, or as an RNP complex, in multiple cell types.
5. Anti-CRISPR Regulation
To date, five families of Acrs from diverse bacterial species have been shown to inhibit Nme1Cas9 in vitro and in human cells (Pawluk et al., 2016; Lee et al., 2018, submitted).
Considering the high sequence identity between Nme1Cas9 and Nme2Cas9, at least some of these Acr families should inhibit Nme2Cas9. To test this, all five families of recombinant Acrs were expressed, purified and tested for Nme2Cas9’s ability to cleave a target in vitro in the presence of a member of each family (10:1 Acr:Cas9 molar ratio). An inhibitor was used for the type I-E CRISPR system in E. coli (AcrE2) as a negative control, while Nme1Cas9was used as a positive control. (Pawluk et al., 2014); (Pawluk et al., 2016). As expected, all 5 families inhibited Nme1Cas9, while AcrE2 failed to do so. See, Figure 11A, top. AcrIIC1Nme, AcrIIC2Nme,
AcrIIC3Nme, and AcrIIC4Hpa completely inhibited Nme2Cas9. Strikingly, however, AcrIIC5Smu which has been previously reported as the most potent of the Nme1Cas9 inhibitors (Lee et al., 2018), did not inhibit Nme2Cas9 in vitro even at a 10-fold molar excess. This suggests that it likely inhibits Nme1Cas9 by interacting with its PID.
Figure 10 presents exemplary data showing dose dependence and segmental deletions by Nme2Cas9, as related to Figure 9. Figure 10A shows exemplary increasing the dose of electroporated Nme2Cas9 plasmid (500 ng, vs.200 ng in Figure 3A) improves editing efficiency at two sites (TS16 and TS6). Data provided in yellow are re-used from Figure 9A. Figure 10B shows exemplary Nme2Cas9 can be used to create precise segmental deletions. Two TLR2.0 targets with cleavage sites 32 bp apart were targeted simultaneously with Nme2Cas9. The majority of lesions created were deletions of exactly 32 bp (blue).
Figure 11 presents exemplary data showing that Nme2Cas9 is subject to inhibition by a subset of type II-C anti-CRISPR families in vitro and in cells. All experiments were done in triplicate and error bars represent s.e.m. Figure 11A shows exemplary In vitro cleavage assay of Nme1Cas9 and Nme2Cas9 in the presence of five previously characterized anti-CRISPR proteins (10:1 ratio of Acr:Cas9). Top: Nme1Cas9 efficiently cleaves a fragment containing a protospacer with an N4GATT PAM in the absence of an Acr or in the presence of a negative control Acr (AcrE2). All five previously characterized type II-C Acr families inhibited
Nme1Cas9, as expected. Bottom: Nme2Cas9 inhibition mirrors that of Nme1Cas9, except for the lack of inhibition by AcrIIC5Smu. Figure 11B shows exemplary genome editing in the presence of the five previously described anti-CRISPR families. Plasmids expressing Nme2Cas9 (200 ng), sgRNA (100 ng) and each respective Acr (200 ng) were co-transfected into HEK293T cells, and genome editing was measured using Tracking of Indels by Decompostion (TIDE) 72 hr post transfection. Consistent with our in vitro analyses, all type II-C anti-CRISPRs except AcrIIC5Smu inhibited genome editing, albeit with different efficiencies. Figure 11C shows exemplary Acr inhibition of Nme2Cas9 is dose-dependent with distinct apparent potencies. Nme2Cas9 is fully inhibited by AcrIIC1Nme and AcrIIC4Hpa at 2:1 and 1:1 mass ratios of cotransfected Acr and Nme2Cas9 plasmids, respectively.
To further test this, a Nme1Cas9/Nme2Cas9 chimera with the PID of Nme2Cas9 was tested. See, Figure 5D and Figure 6D. Due to the reduced activity of this hybrid, a ~30x higher concentration of Cas9 was used to achieve a similar cleavage efficiency while maintaining the 10:1 Cas9:Acr molar ratio. No inhibition was observed by AcrIIC5Smu on this protein chimera. See, Figure 12. This data provides further evidence that AcrIIC5Smu likely interacts with the PID of Nme1Cas9. Regardless of the mechanistic basis for the differential inhibition by AcrIIC5Smu, these results indicate that Nme2Cas9 is subject to inhibition by the other four type II-C Acr families.
Figure 12 presents exemplary data showing that a Nme2Cas9 PID swap renders
Nme1Cas9 insensitive to AcrIIC5Smu inhibition, as related to Figure 11. In vitro cleavage by the Nme1Cas9-Nme2Cas9PID chimera in the presence of previously characterized Acr proteins (10 uM Cas9-sgRNA + 100 uM Acr).
Based on the above in vitro data, it was hypothesized that AcrIIC1Nme, AcrIIC2Nme, AcrIIC3Nme, and AcrIIC4Hpa could be used as off-switches for Nme2Cas9 genome editing. To test this, Nme2Cas9/sgRNA plasmid transfections (150 ng of each plasmid) targeting TS16 were performed in HEK293T cells in the presence or absence of Acr expression plasmids, as it has been reported that most Acrs inhibited Nme1Cas9 at those plasmid ratios (Pawluk et al., 2016). As expected, AcrIIC1Nme, AcrIIC2Nme, AcrIIC3Nme and AcrIIC4Hpa inhibited Nme2Cas9 genome editing, while AcrIIC5Smu had no effect. See, Figure 11B. Complete inhibition was observed by AcrIIC3Nme and AcrIIC4Hpa, suggesting that they have high potency against Nme2Cas9 as compared to AcrIIC1Nme and AcrIIC2Nme. To further compare the potency of AcrIIC1Nme and AcrIIC4Hpa, we repeated the experiments at various ratios of Acr plasmid to Cas9 plasmid. See, Figure 11C. The data show that the AcrIIC4Hpa plasmid is especially potent against Nme2Cas9. Together, these data suggest that several Acr proteins can be used as off-switches for
Nme2Cas9-based applications.
6. Hyper-Accuracy
Nme1Cas9 demonstrates remarkable editing fidelity in cells and mouse models (Lee et al., 2016; Amrani et al., 2018; Ibraheim et al., 2018). Furthermore, the similarity of Nme2Cas9 to Nme1Cas9 over most of its length suggests that it may likewise be hyper-accurate. However, the higher number of sites sampled in the genome as a result of the dinucleotide PAM could create more opportunities for Nme2Cas9 off-targeting in comparison with Nme1Cas9 and its less frequently encountered 4-nucleotide PAM. To assess the off-target profile of Nme2Cas9, GUIDE-seq (genome-wide, unbiased identification of double-stranded breaks enabled by sequencing) was used to identify potential off-target sites empirically and in an unbiased fashion (Tsai et al., 2014). Even the best off-target prediction algorithms are prone to false negatives necessitating empirical target site profiling methods (Bolukbasi et al., 2015b; Tsai and Joung, 2016; Tycko et al., 2016). GUIDE-seq relies on the incorporation of double-stranded oligodeoxynucleotides (dsODNs) into DNA double-stranded break sites throughout the genome. These insertion sites are then detected by amplification and high-throughput sequencing.
Because SpyCas9 is a well-characterized Cas9 ortholog it is useful for multiplexed applications with other Cas9s, and as a benchmark for their editing properties (Jiang and Doudna, 2017; Komor et al., 2017). SpyCas9 and Nme2Cas9 were cloned into identical plasmid backbones, with the same UTRs, linkers, NLSs, and promoters, for parallel transient transfections (along with similarly matched sgRNA-expressing plasmids) into HEK293T cells. First, it was confirmed that the RNA guides for SpyCas9 and Nme2Cas9 are orthogonal, i.e. that Nme2Cas9 sgRNAs do not direct editing by SpyCas9, and vice versa. See, Figure 13A. This was in contrast to earlier reported results with Nme1Cas9 (Esvelt et al., 2013; Fonfara et al., 2014).
Next, to identify a use of SpyCas9 as a benchmark for GUIDE-seq, because SpyCas9 and Nme2Cas9 have non-overlapping PAMs its can therefore potentially edit any dual site (DS) flanked by a 5’-NGGNCC-3’ sequence, which simultaneously fulfills the PAM requirements of both Cas9’s. This permits side-by-side comparisons of off-targeting with RNA guides that facilitate an edit of the exact same on-target site. See, Figure 14A. Six (6) DSs in VEGFA were targeted, each of which also has a G at the appropriate positions 5’ of the PAM such that both SpyCas9 and Nme2Cas9 guides (driven by the U6 promoter) were 100% complementary to the target site. Seventy-two (72) hours after transfection, a TIDE analysis was performed on these sites targeted by each nuclease. Nme2Cas9 induced indels at all six sites, albeit at low efficiencies at two of them, while SpyCas9 induced indels at four of the six sites. See, Figure 14B. At two of the four sites (DS1 and DS4) at which SpyCas9 was effective, it induced ~7-fold more indels than Nme2Cas9, while Nme2Cas9 induced a ~3-fold higher frequency of indels than SpyCas9 at DS6. Both Cas9 orthologs edited DS2 with approximately equal efficiency.
For GUIDE-seq, DS2, DS4 and DS6 were selected to sample off-target cleavage with Nme2Cas9 guides that direct on-target editing as efficiently, less efficiently, or more efficiently than the corresponding SpyCas9 guides, respectively. In addition to the three dual sites, TS6 was added as it has been observed to be an efficiently edited Nme2Cas9 target sites, having an approximate 30-50% indel efficiency depending on the cell type. See, Figures 9A and 10A. Similar data is seen with the mouse Pcsk9 and Rosa26 Nme2Cas9 sites. See, Figure 9B.
Plasmid transfections were performed for each Cas9 along with their cognate sgRNAs and the dsODNs. Subsequently, GUIDE-seq libraries were prepared as described previously (Amrani et al., 2018). A GUIDE-seq analysis revealed efficient on-target editing for both Cas9 orthologs, with relative efficiencies (as reflected by GUIDE-seq read counts) that are similar to those observed by TIDE. Figure 13B and Table 2. (Tsai et al., 2014; Zhu et al., 2017).
Figure 13 presents exemplary data showing orthogonality and relative accuracy of Nme2Cas9 and SpyCas9 at dual target sites, as related to Figure 12. Figure 13A shows exemplary Nme2Cas9 and SpyCas9 guides are orthogonal. TIDE results show the frequencies of indels created by both nucleases targeting DS2 with either their cognate sgRNAs or with the sgRNAs of the other ortholog. Figure 13B shows exemplary Nme2Cas9 and SpyCas9 exhibiting comparable on-target editing efficiencies as assessed by GUIDE-seq. Bars indicate on-target read counts from GUIDE-Seq at the three dual sites targeted by each ortholog. Orange bars represent Nme2Cas9 and black bars represent SpyCas9. Figure 13C shows an exemplary SpyCas9’s on- target vs. off-target read counts for each site. Orange bars represent the on-target reads while black bars represent off-targets. Figure 13D shows exemplary Nme2Cas9’s on-target vs. off- target reads for each site. Figure 13E bar graphs showing exemplary indel efficiencies (measured by TIDE) at potential off-target sites predicted by CRISPRSeek. On- and off-target site sequences are shown on the left, with the PAM region underlined and sgRNA mismatches and non-consensus PAM nucleotides given in red. Table 2: GUIDE-seq Data
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
Figure imgf000060_0001
Figure imgf000061_0001
Figure imgf000062_0001
For off-target identification, the analysis revealed that the DS2, DS4, and DS6
SpyCas9 sgRNAs appeared to direct editing at 93, 10, and 118 candidate off-target sites, respectively, in the normal range of off-targets when plasmid-based SpyCas9 editing is analyzed by GUIDE-seq (Fu et al., 2014; Tsai et al., 2014). In striking contrast, the DS2, DS4, and DS6 Nme2Cas9 sgRNAs appeared to direct editing at 1, 0, and 1 off-target sites, respectively. Figure 14C and Table 2. When compared to the GUIDE-seq read counts for the SpyCas9 off-targets, those of Nme2Cas9 were very low, further suggesting that Nme2Cas9 is highly specific. Figure 13C cf. Figure 13D. Nme2Cas9 GUIDE-seq analyses with the TS6, Pcsk9, and Rosa26 yielded similar results (0, 0, and 1 off-target sites, respectively, with a modest read count for the Rosa26-OT1 off-target site). Figure 13C, Figure 14D, and Table 2.
Figure 14 presents exemplary data showing that Nme2Cas9 exhibits little or no detectable off-targeting in mammalian cells. Figure 14Ashows an exemplary schematic depicting dual sites (DSs) targetable by both SpyCas9 and Nme2Cas9 by virtue of their non-overlapping PAMs. The Nme2Cas9 PAM (orange) and SpyCas9 PAM (blue) are highlighted. A 24nt Nme2Cas9 guide sequence is indicated in yellow; the corresponding guide sequence for SpyCas9 would be 4nt shorter at the 5’ end. Figure 14B shows an exemplary Nme2Cas9 and SpyCas9 that both induce indels at DSs. Six DSs in VEGFA (with GN3GN19NGGNCC sequences) were selected for direct comparisons of editing by the two orthologs. Plasmids expressing each Cas9 (with the same promoter, linkers, tags and NLSs) and its cognate guide were transfected into HEK293T cells. Indel efficiencies were determined by TIDE 72 hrs post transfection. Nme2Cas9 editing was detectable at all six sites and was marginally or significantly more efficient than SpyCas9 at two sites (DS2 and DS6, respectively). SpyCas9 edited four out of the six sites (DS1, DS2, DS4 and DS6), with two sites showing significantly higher editing efficiencies than
Nme2Cas9 (DS1 and DS4). DS2, DS4 and DS6 were selected for GUIDE-Seq analysis as Nme2Cas9 was equally efficient, less efficient and more efficient than SpyCas9, respectively, at these sites. Figure 14C shows exemplary Nme2Cas9 genome editing that is highly accurate in human cells. Numbers of off-target sites detected by GUIDE-Seq for each nuclease at individual target sites are shown. In addition to dual sites, we analyzed TS6 (because of its high on-target editing efficiency) and Pcsk9 and Rosa26 sites in mouse Hepa1-6 cells (to measure accuracy in another cell type). Figure 14D shows an exemplary targeted deep sequencing to detect indels in edited cells confirms the high Nme2Cas9 accuracy indicated by GUIDE-seq. Figure 14E shows an exemplary sequence for the validated off-target site of the Rosa26 guide, showing the PAM region (underlined), the consensus CC PAM dinucleotide (bold), and three mismatches in the PAM-distal portion of the spacer (red).
To validate the off-target sites detected by GUIDE-seq, a targeted deep sequencing was performed to measure indel formation at the top off-target loci following GUIDE-seq- independent editing (i.e. without co-transfection of the dsODN). While SpyCas9 showed considerable editing at most off-target sites tested and, in some instances, was more efficient than that at the corresponding on-target site, Nme2Cas9 exhibited no detectable indels at the lone DS2 and DS6 candidate off-target sites. See, Figure 14D. With the Rosa26 sgRNA, Nme2Cas9 induced ~1% editing at the Rosa26-OT1 site in Hepa1-6 cells, compared to ~30% on-target editing. See, Figure 14D. It is noteworthy that this off-target site has a consensus Nme2Cas9 PAM (ACTCCCT) with only 3 mismatches at the PAM-distal end of the guide- complementary region (i.e. outside of the seed). See, Figure 14E. These data support and reinforce our GUIDE-seq results indicating a high degree of accuracy for Nme2Cas9 genome editing in mammalian cells.
To further corroborate the above GUIDE-Seq results, CRISPRseek was used to computationally predict potential off-target sites for two active Nme2Cas9 sgRNAs that targeted TS25 and TS47, both of which are also in VEGFA See, Figure 9A; (Zhu et al., 2014). Three (TS25) or four (TS47) of the most closely matched predicted sites, five with N4CC PAMs and two with N4CA PAMs; each had 2-5 mismatches, mostly in their PAM-distal, non-seed regions. See, Figure 13E. On- vs. off-target editing was compared after Nme2Cas9+sgRNA plasmid transfections into HEK293T cells by targeted amplification of each locus, followed by TIDE analysis. Consistently, no indels could be detected at those off-target sites for either sgRNA by TIDE, while efficient on-target editing was readily detected in DNA from the same populations of cells. Taken together, our data indicate that Nme2Cas9 is a naturally hyper-accurate genome editing platform in mammalian cells.
7. Associated Adenovirus Delivery
The compact size, small PAM, and high fidelity of Nme2Cas9 offer major advantages for in vivo genome editing using Associated Adenovirus (AAV) delivery. To test whether effective Nme2Cas9 genome editing can be achieved via single-AAV delivery, Nme2Cas9 was cloned with its sgRNA and their promoters (U1a and U6, respectively) into an AAV vector backbone. See, Figure 15A. An all-in-one AAV was prepared with an sgRN- .Nme2Cas9 packaged into a hepatotropic AAV8 capsid to target two genes in the mouse liver: i) Rosa26 (a commonly used safe harbor locus for transgene insertion) (Friedrich and Soriano, 1991) as a negative control; and ii) Pcsk9, a major regulator of circulating cholesterol homeostasis (Rashid et al., 2005), as a phenotypic target.
SauCas9- or Nme1Cas9-induced indels in Pcsk9 in the mouse liver results and reduced cholesterol levels providing a useful and easy-to-score in vivo benchmark for new editing platforms (Ran et al., 2015; Ibraheim et al., 2018). The Nme2Cas9 RNA guides were the same as those used above. See, Figure 9B, Figure 13D, and Figure 14. As Rosa26-OT1 was the only Nme2Cas9 off-target site that has been validated in cultured mammalian cells, the Rosa26 guide also provided us with an opportunity to assess on- vs. off-target editing in vivo. See, Figures 14D-E). The tail veins of two groups of mice (n = 5) were injected with 4 x 1011 AAV8.sgRNA.Nme2Cas9 genome copies (GCs) targeting either Pcsk9 or Rosa26. Serum was collected at 0, 14 and 28 days post-injection for cholesterol level measurement. Mice were sacrificed at 28 days post-injection and liver tissues were harvested. See, Figure 15A.
Targeted deep sequencing of each locus revealed ~38% and ~46% indel induction at the Pcsk9 and Rosa26 editing sites, respectively, in the liver. See, Figure 15B. Because hepatocytes constitute only 65-70% of total cellular content in the adult liver, Nme2Cas9 AAV-induced hepatocyte editing efficiencies with sgPcsk9 and sgRosa were approximately 54-58% and 66-71%, respectively (Racanelli and Rehermann, 2006).
Only 2.25% liver indels overall (~3-3.5% in hepatocytes) were detected at the Rosa26- OT1 off-target site, comparable to the 1% editing that we observed at this site in transfected Hepa1-6 cells. Figure 15B cf Figure 14D. At both 14 and 28 days post-injection, Pcsk9 editing was accompanied by a ~44% reduction in serum cholesterol levels, whereas mice treated with the sgRosa26-expressing AAV maintained normal level of cholesterol throughout the study. See, Figure 15C. The ~44% reduction in serum cholesterol in the Nme2Cas9/sgPcsk9 AAV- treated mice compares well with the ~40% reduction reported with SauCas9 all-in-one AAV when targeting the same gene (Ran et al., 2015).
Figure 15 presents exemplary data showing Nme2Cas9 genome editing in vivo via all-in- one AAV delivery. Figure 15A shows exemplary workflow for delivery of
AAV8.sgRNA.Nme2Cas9 to lower cholesterol levels in mice by targeting Pcsk9. Top: schematic of the all-in-one AAV vector expressing Nme2Cas9 and the sgRNA (individual genome elements not to scale). BGH, bovine growth hormone poly(A) site; HA, epitope tag; NLS, nuclear localization sequence; h, human-codon-optimized. Bottom: Timeline for
AAV8.sgRNA.Nme2Cas9 tail-vein injections (4 x 1011 GCs), followed by cholesterol measurements at day 14 and indel, histology and cholesterol analyses at day 28 post-injection. Figure 15B shows an exemplary TIDE analysis to measure indels in DNA extracted from livers of mice injected with AAV8.Nme2Cas9+sgRNA targeting Pcsk9 and Rosa26 (control) loci. Indel efficiency at the lone off-target site identified by GUIDE-seq for these two sgRNAs (Rosa26|OT1) were also assessed by TIDE. Figure 15C shows an exemplary reduced serum cholesterol levels in mice injected with the Pcsk9-targeting guide compared to the Rosa26- targeting controls. P values are calculated by unpaired two-tailed t-test. Figure 16 presents exemplary data showing PCSK9 knockdown and liver histology following Nme2Cas9 AAV delivery and editing, related to Figure 15. Figure 16A shows exemplary Western blotting using anti-PCSK9 antibody reveals strongly reduced levels of PCSK9 in the livers of mice treated with sgPcsk9, compared to mice treated with sgRosa26.2ng of recombinant PCSK9 was used as a mobility standard (left-most lane), and a cross-reacting band in the liver samples is indicated by an asterisk. GAPDH was used as loading control (bottom panel). Figure 16B shows exemplary H&E staining from livers of mice injected with AAV8.Nme2Cas9+sgRosa26 (left) or
AAV8.Nme2Cas9+sgPcsk9 (right) vectors. Scale bars, 25 mm.
Western blotting was performed using an anti-PCSK9 antibody to estimate PCSK9 protein levels in the livers of mice treated with sgPcsk9 and sgRosa26. Liver PCSK9 was below the detection limit in mice treated with sgPcsk9, whereas sgRosa26-treated mice exhibited normal levels of PCSK9. See, Figure 16A. Hematoxylin and eosin (H&E) staining and histology revealed no signs of toxicity or tissue damage in either group after Nme2Cas9 expression. See, Figure 16B. These data validate Nme2Cas9 as a highly effective genome editing system in vivo, including when delivered by single-AAV vectors.
AAV vectors have recently been used for the generation of genome-edited mice, without the need for microinjection or electroporation, simply by soaking the zygotes in culture medium containing AAV vector(s), followed by reimplantation into pseudopregnant females (Yoon et al., 2018). Editing was obtained previously with a dual-AAV system in which SpyCas9 and its sgRNA were delivered in separate vectors (Yoon et al., 2018). To test whether Nme2Cas9 could perform accurate and efficient editing in mouse zygotes with an all - in-one AAV delivery system, we targeted Tyrosinase (Tyr). A bi-allelic inactivation of Tyr disrupts melanin production resulting in an albino phenotype (Yokoyama et al., 1990).
An efficient Tyr sgRNA was validated that cleaves the Tyr locus only seventeen (17) bp from the site of the classic albino mutation in Hepa1-6 cells by transient transfections. See, Figure 17A. Next, C57BL/6NJ zygotes were incubated for 5-6 hours in culture medium containing 3x109 or 3x108 GCs of an all-in-one AAV6 vector expressing Nme2Cas9 along with the Tyr sgRNA. After overnight culture in fresh media, those zygotes that advanced to the two-cell stage were transferred to the oviduct of pseudopregnant recipients and allowed to develop to term. See, Figure 18A. Coat color analysis of pups revealed mice that were albino, chinchilla (indicating a hypomorphic allele of Tyrosinase), or that had variegated coat color composed of albino and chinchilla spots but lacking black pigmentation. See, Figures 18B-C. These results suggest a high frequency of biallelic mutations since the presence of a wild-type Tyrosinase allele should render black pigmentation. A total of five pups (10%) were born from the 3x109 GCs experiment. All of them carried indels; phenotypically, two were albino, one was chinchilla, and two had variegated pigmentation, indicating mosaicism.
From the 3x108 GCs experiment, four (4) pups (14%) were obtained, two of which died at birth, preventing a coat color or genome analysis. Coat color analysis of the remaining two pups revealed one chinchilla and one mosaic pup. These results indicate that single-AAV delivery of Nme2Cas9 and its guide can be used to generate mutations in mouse zygotes without microinjection or electroporation.
To measure on-target indel formation in the Tyr gene, DNA was isolated from the tails of each mouse, the locus was amplified and upon which a TIDE analysis was performed. All mice had high levels of on-target editing by Nme2Cas9, varying from 84% to 100%. See, Figures 17B-C. Most lesions in albino mouse 9-1 were either a 1- or a 4-bp deletion, suggesting either mosaicism or trans-heterozygosity, but albino mouse 9-2 exhibited a uniform 2-bp deletion. See, Figure 17C. Figure 17 presents exemplary data showing Tyr editing ex vivo in mouse zygotes, related to Figure 16. Figure 17A shows an exemplary two sites in Tyr, each with N4CC PAMs, were tested for editing in Hepa1-6 cells. The sgTyr2 guide exhibited higher editing efficiency and was selected for further testing. Figure 17B shows an exemplary seven mice that survived post-natal development, and each exhibited coat color phenotypes as well as on-target editing, as assayed by TIDE. Figure 17C shows an exemplary Indel spectra from tail DNA of each mouse from (B), as well as an unedited C57BL/6NJ mouse, as indicated by TIDE analysis. Efficiencies of insertions (positive) and deletions (negative) of various sizes are indicated.
Figure 18 presents exemplary data showing Nme2Cas9 genome editing ex vivo via all-in- one AAV delivery. Figure 18A shows an exemplary workflow for single-AAV Nme2Cas9 editing ex vivo to generate albino C57BL/6NJ mice by targeting the Tyr gene. Zygotes are cultured in KSOM containing AAV6.Nme2Cas9:sgTyr for 5-6 hours, rinsed in M2, and cultured for a day before being transferred to the oviduct of pseudo-pregnant recipients. Figure 18B shows exemplary albino (left) and chinchilla or variegated (middle) mice generated by 3x109 GCs, and chinchilla or variegated mice (right) generated by 3x108 GCs of zygotes with
AAV6.Nme2Cas9:sgTyr. Figure 18C shows an exemplary summary of Nme2Cas9.sgTyr single- AAV ex vivo Tyr editing experiments at two AAV doses.
The data is inconclusive as to whether there was no mosaicism in mouse 9-2, or that additional alleles were absent from mouse 9-1, because only tail samples were sequenced and other tissues could have distinct lesions. Analysis of tail DNA from chinchilla mice revealed the presence of in-frame mutations that are potentially the cause of the chinchilla coat color. The limited mutational complexity suggests that editing occurred early during embryonic development in these mice. These results provide a streamlined route toward mammalian mutagenesis through the application of a single AAV vector, in this case delivering both Nme2Cas9 and its sgRNA.
Figure 19 shows an exemplary mCherry reporter assay for nSpCas9-ABEmax and optimized nNme2Cas9-ABEmax activities. Figure 19A shows exemplary sequence information of ABE-mCherry reporter. There is a TAG stop codon in the mCherry coding region. In the reporter-integrated stable cell line, there is no mCherry signal due to this stop codon. The mCherry signal will be activated if the nSpCas9-ABEmax or optimized nNme2Cas9-ABEmax can convert TAG to CAG, which encodes a glutamine residue. Figure 19B shows an exemplary mCherry signal is activated due to SpCas9-ABE or Nme2Cas9-ABE activity. Upper panel:
negative control (no editing); middle panel: mCherry activation by nSpCas9-ABEmax; bottom panel: mCherry activation by optimized nNme2Cas9-ABEmax. Figure 19C shows an exemplary FACS quantitation of base editing events in mCherry reporter cells transfected with the SpCas9- ABE or Nme2Cas9-ABE. N = 6; error bars represent S.D. Results are from three biological replicates performed in technical duplicates.
Figure 20 shows an exemplary GFP reporter assay for nSpCas9-CBE4 (Addgene #100802) and nNme2Cas9-CBE4 (same plasmid backbone as Addgene #100802) activities. Figure 20A shows exemplary sequence information of the CBE-GFP reporter. There is a mutation that converts GYG to GHG in the fluorophore core region of the GFP reporter line. There is no GFP signal due to this mutation. The GFP signal will be activated if the nSpCas9- CBE4 or nNme2Cas9-CBE4 can convert CAC (encoding histidine) to TAC/TAT (encoding tyrosine). Figure 20B shows an exemplary GFP signal is activated due to nSpCas9-CBE4 or nNme2Cas9-CBE4 activity. Upper panel: negative control (no editing); middle panel: GFP activation by nSpCas9-CBE4; bottom panel: GFP activation by nNme2Cas9-CBE4). Figure 20C shows an exemplary FACS quantitation of base editing events in GFP reporter cells transfected with nSpCas9-CBE4 or nNme2Cas9-CBE4. N = 6; error bars represent S.D. Results are from biological replicates performed in technical duplicates.
Figure 21 shows exemplary cytosine editing by nNme2Cas9-CBE4. Upper panel shows the KANK3 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples. Bottom panel shows the quantification of the substitution efficiency of each type of base in the nNmeCas9-CBE4 editing window of the KANK3 target sequences. Sequence tables show nucleotide frequencies at each position.
Frequencies of expected C-to-T conversion are indicated in red.
Figure 22 shows exemplary cytosine and adenine editing by nNme2Cas9-CBE4 and
nNme2Cas9-ABEmax, respectively. Upper panel shows the PLXNB2 targeting sequence information (PAM sequences are indicated in red) of Nme2Cas9 and base editing in the negative control samples. Middle panel shows the quantification of the substitution rate of each type of base in the nNmeCas9-ABEmax editing windows of the PLXNB2 target sequence. Sequence tables show nucleotide frequencies at each position. Frequencies of expected A-to-G conversion are highlighted in red. Bottom panel shows the quantification of the substitution efficiency of each type of base in the nNmeCas9-CBE4 editing windows of the PLXNB2 target sequence. Sequence tables show nucleotide frequencies at each position. Frequencies of expected C-to-T conversion are highlighted in red.
8. Sequences Alignment of Nme1Cas9 and Nme2Cas9
Non-PID aa differences (teal- underlined); PID aa differences (yellow - underlined bold); active site residues (red - bold). Nme1Cas9 (1-60)
MAAFKPNSINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAM
Nme2Cas9 (1-60)
MAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAM Nme1Cas9 (61-120)
ARRLARSVRRLTRRRAHRLLRTRRLLKREGVLQAANFDENGLIKSLPNTPWQLRAAAL DR
Nme2Cas9 (61-120)
ARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAAL DR Nme1Cas9 (121-180)
KLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVAGNAHALQTGDFRTPA EL
Nme2Cas9 (121-180)
KLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPA EL Nme1Cas9 (181-240)
ALNKFEKESGHIRNQRSDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLM
Nme2Cas9 (181-240)
ALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLM Nme1Cas9 (241-300)
TQRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTD T Nme2Cas9 (241-300)
TQRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTD T Nme1Cas9 (301-360)
ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISR AL
Nme2Cas9 (301-360)
ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISR AL Nme1Cas9 (361-420)
EKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLKDRIQPEILEALLKHISFDKF
Nme2Cas9 (361-420)
EKEGLKDKKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKF Nme1Cas9 (421-480)
VQISLKALRRIVPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRA
Nme2Cas9 (421-480)
VQISLKALRRIVPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRA Nme1Cas9 (481-540)
LSQARKVINGVVRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFRE Y
Nme2Cas9 (481-540)
LSQARKVINGVVRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFRE Y Nme1Cas9 (541-600)
FPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSF Nme2Cas9 (541-600)
FPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSF Nme1Cas9 (601-660)
NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFD ED
Nme2Cas9 (601-660)
NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFD ED Nme1Cas9 (661-720)
GFKERNLNDTRYVNRFLCQFVADRMRLTGKGKKRVFASNGQITNLLRGFWGLRKVRA END
Nme2Cas9 (661-720)
GFKECNLNDTRYVNRFLCQFVADHILLTGKGKRRVFASNGQITNLLRGFWGLRKVRAE ND Nme1Cas9 (721-780)
RHHALDAVVVACSTVAMQQKITRFVRYKEMNAFDGKTIDKETGEVLHQKTHFPQPWE FFA
Nme2Cas9 (721-780)
RHHALDAVVVACSTVAMQQKITRFVRYKEMNAFDGKTIDKETGKVLHQKTHFPQPWE FFA Nme1Cas9 (781-840)
QEVMIRVFGKPDGKPEFEEADTLEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMS G
Nme2Cas9 (781-840)
QEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMS G Nme1Cas9 (841-895)
QGHMETVKSAK---RLDEGVSVLRVPLTQLKLKDLEKMVNR-- EREPKLYEALKARLEAH
Nme2Cas9 (841-899)
AHK- DTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMVNYKNGREIELYEALKARLEAY Nme1Cas9 (896-950)
KDDPAKAFAE---PFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNH-- NGIADNATMVRV
Nme2Cas9 (900–954)
GGNAKQAFDPKDNPFYKK---G-- GQLVKAVRVEKTQESGVLLNKKNAYTIADNGDMVRV Nme1Cas9 (951-1005)
DVFEKG----- DKYYLVPIYSWQVAKGILPDRAVVQGKDEEDWQLIDDSFNFKFSLHPND
Nme2Cas9 (955-1007)
DVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKG-------YRIDDSYTFCFSLHKYD Nme1Cas9 (1006-1063)
LVEVIT-- KKARMFGYFASCHRGTGNINIRIHDLDHKIGKNGILEGIGVKTALSFQKYQI
Nme2Cas9 (1008-1063)
LIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQ---- QFRISTQNLVLIQKYQV Nme1Cas9 (1064-1082)
DELGKEIRPCRLKKRPPVR
Nme2Cas9 (1064-1082)
NELGKEIRPCRLKKRPPVR Alignment of Nme1Cas9 and Nme3Cas9
Non-PID aa differences (teal- underlined); PID aa differences (yellow - underlined bold); active site residues (red - bold). Nme1Cas91 MAAFKPNSINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAE 50 Nme3Cas91 MAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAE 50 Nme1Cas951
VPKTGDSLAMARRLARSVRRLTRRRAHRLLRTRRLLKREGVLQAANFDEN 100 Nme3Cas951
VPKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDEN 100 Nme1Cas9101
GLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGET 150
Nme3Cas9101
GLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGET 150 Nme1Cas9151
ADKELGALLKGVAGNAHALQTGDFRTPAELALNKFEKESGHIRNQRSDYS 200 Nme3Cas9151
ADKELGALLKGVADNAHALQTGDFRTPAELALNKFEKECGHIRNQRGDYS 200 Nme1Cas9201 HTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA 250
Nme3Cas9201
HTFSRKDLQAELNLLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDA 250 Nme1Cas9251
VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDT 300 Nme3Cas9251
VQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDT 300 Nme1Cas9301
ERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEM 350 Nme3Cas9301
ERATLMDEPYRKSKLTYAQARKLLSLEDTAFFKGLRYGKDNAEASTLMEM 350 Nme1Cas9351 KAYHAISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK 400
Nme3Cas9351 KAYHTISRALEKEGLKDKKSPLNLSPELQDEIGTAFSLFKTDEDITGRLK 400 Nme1Cas9401 DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG 450
Nme3Cas9401 DRIQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYG 450 Nme1Cas9451 DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR 500
Nme3Cas9451 DHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPAR 500 Nme1Cas9501 IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS 550
Nme3Cas9501 IHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKS 550 Nme1Cas9551
KDILKLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSF 600 Nme3Cas9551
KDILKLRLYEQQHGKCLYSGKEINLGRLNEKGYVEIDHALPFSRTWDDSF 600 Nme1Cas9601
NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ 650 Nme3Cas9601
NNKVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQ 650 Nme1Cas9651
RILLQKFDEDGFKERNLNDTRYVNRFLCQFVADRMRLTGKGKKRVFASNG 700 Nme3Cas9651
RILLQKFDEDGFKERNLNDTRYVNRFLCQFVADRMRLTGKGKKRVFASNG 700 Nme1Cas9701
QITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEM 750 Nme3Cas9701
QITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEM 750 Nme1Cas9751
NAFDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEA 800 Nme3Cas9751
NAFDGKTIDKETGEVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEA 800 Nme1Cas9801
DTLEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGQGHMETVKSA 850 Nme3Cas9801
DTPEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGQGHMETVKSA 850 Nme1Cas9851
KRLDEGVSVLRVPLTQLKLKDLEKMVNREREPKLYEALKARLEAHKDDPA 900 Nme3Cas9851
KRLDEGVSVLRVPLTQLKLKDLEKMVNREREPKLYEALKARLEAHKDDPA 900 Nme1Cas9
901 KAFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNHNGIADNATMVRV 950 Nme3Cas9
901 KAFAEPFYKYDKAGNRTQQVKAVRVEQVQKTGVWVRNHNGIADNATMVRV 950 Nme1Cas9951
DVFEKGDKYYLVPIYSWQVAKGILPDRAVVQGKDEEDWQLIDDSFNFKFS 1000 Nme3Cas9951
DVFEKGDKYYLVPIYSWQVAKGILPDRAVVAYADEEDWTVIDESFRFKFV 1000 Nme1Cas91001
LHPNDLVEVITKKARMFGYFASCHRGTGNINIRIHDLDHKIGKNGILEGI 1050 Nme3Cas91001 LYSNDLIKVQLKKDSFLGYFSGLDRATGAISLREHDLEKSKGKDG- MHRI 1049 Nme1Cas91051 GVKTALSFQKYQIDELGKEIRPCRLKKRPPVR 1082
Nme3Cas91050 GVKTALSFQKYQIDEMGKEIRPCRLKKRPPVR 1081 Plasmid-Expressed Nme2Cas9
SV40 NLS (yellow- BOLD); 3X-HA-Tag (green-(underlined/bold); cMyc-like NLS (teal- plain); Linker (magenta - bold italics) and Nme2Cas9 (italics). MAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKTGDSLAMARRL ARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAALDRKLTPLEW SAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAELALNKFEKES GHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQRPALSGDAV QKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATLMDEPYRKSK LTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKDKKSPLNLSSE LQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVPLMEQGKRY DEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGSPARIHIETA REVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQQHGKCLY SGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYFNGKDNSR EWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHILLTGKGK RRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRYKEMNAFD GKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRTLLAEKLSS RPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLADLENMVN YKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQESGVLLNKKN AYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDDSYTFCFSLH KYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQKYQVNELG KEIRPCRLKKRPPVRGTGGPKKKRKVYPYDVPDYAGYPYDVPDYAGSYPYDVPDYAG SAAPAAKKKKLDFESG* AAV-expressed Nme2Cas9
SV40 NLS (yellow- BOLD); 3X-HA-Tag (green-(underlined/bold); Nucleoplasmin-like NLS (red-underline); c-myc NLS (teal- plain); Linker (magenta - bold italics) and Nme2Cas9 (italics). MVPKKKRKVEDKRPAATKKAGQAKKKKMAAFKPNPINYILGLDIGIASVGWAMVEIDEE ENPIRLIDLGVRVFERAEVPKTGDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAAD FDENGLIKSLPNTPWQLRAAALDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALL KGVANNAHALQTGDFRTPAELALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKE FGNPHVSGGLKEGIETLLMTQRPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKL NNLRILEQGSERPLTDTERATLMDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEAST LMEMKAYHAISRALEKEGLKDKKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEA LLKHISFDKFVQISLKALRRIVPLMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNP VVLRALSQARKVINGVVRRYGSPARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFRE YFPNFVGEPKSKDILKLRLYEQQHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNN KVLVLGSENQNKGNQTPYEYFNGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFK ECNLNDTRYVNRFLCQFVADHILLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHAL DAVVVACSTVAMQQKITRFVRYKEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIR VFGKPDGKPEFEEADTPEKLRTLLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSA KRFVKHNEKISVKRVWLTEIKLADLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKD NPFYKKGGQLVKAVRVEKTQESGVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPI YAWQVAENILPDIDCKGYRIDDSYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLA WHDKGSKEQQFRISTQNLVLIQKYQVNELGKEIRPCRLKKRPPVREDKRPAATKKAGQAK KKKYPYDVPDYAGYPYDVPDYAGSYPYDVPDYAAAPAAKKKKLD* Recombinant Nme2Cas9
SV40 NLS (yellow- BOLD); Nucleoplasmin-like NLS (red-underline); Linker (magenta - bold italics) and Nme2Cas9 (italics).
PKKKRKVNAMAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKT GDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAA LDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAE LALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQ RPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATL MDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKD KKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVP LMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGS PARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQ QHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYF NGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHI LLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRY KEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRT LLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLA DLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQES GVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDD SYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQ KYQVNELGKEIRPCRLKKRPPVRGGGGSGGGGSGGGGSPAAKKKKLDGGGSKRPAATK KAGQAKKKK* Recombinant Nme2Cas9 for use in mammalian cell RNP delivery: SV40 NLS (yellow- BOLD); Nucleoplasmin-like NLS (red-underline); Linker (magenta - bold italics) and Nme2Cas9 (italics).
PKKKRKVNAMAAFKPNPINYILGLDIGIASVGWAMVEIDEEENPIRLIDLGVRVFERAEVPKT GDSLAMARRLARSVRRLTRRRAHRLLRARRLLKREGVLQAADFDENGLIKSLPNTPWQLRAAA LDRKLTPLEWSAVLLHLIKHRGYLSQRKNEGETADKELGALLKGVANNAHALQTGDFRTPAE LALNKFEKESGHIRNQRGDYSHTFSRKDLQAELILLFEKQKEFGNPHVSGGLKEGIETLLMTQ RPALSGDAVQKMLGHCTFEPAEPKAAKNTYTAERFIWLTKLNNLRILEQGSERPLTDTERATL MDEPYRKSKLTYAQARKLLGLEDTAFFKGLRYGKDNAEASTLMEMKAYHAISRALEKEGLKD KKSPLNLSSELQDEIGTAFSLFKTDEDITGRLKDRVQPEILEALLKHISFDKFVQISLKALRRIVP LMEQGKRYDEACAEIYGDHYGKKNTEEKIYLPPIPADEIRNPVVLRALSQARKVINGVVRRYGS PARIHIETAREVGKSFKDRKEIEKRQEENRKDREKAAAKFREYFPNFVGEPKSKDILKLRLYEQ QHGKCLYSGKEINLVRLNEKGYVEIDHALPFSRTWDDSFNNKVLVLGSENQNKGNQTPYEYF NGKDNSREWQEFKARVETSRFPRSKKQRILLQKFDEDGFKECNLNDTRYVNRFLCQFVADHI LLTGKGKRRVFASNGQITNLLRGFWGLRKVRAENDRHHALDAVVVACSTVAMQQKITRFVRY KEMNAFDGKTIDKETGKVLHQKTHFPQPWEFFAQEVMIRVFGKPDGKPEFEEADTPEKLRT LLAEKLSSRPEAVHEYVTPLFVSRAPNRKMSGAHKDTLRSAKRFVKHNEKISVKRVWLTEIKLA DLENMVNYKNGREIELYEALKARLEAYGGNAKQAFDPKDNPFYKKGGQLVKAVRVEKTQES GVLLNKKNAYTIADNGDMVRVDVFCKVDKKGKNQYFIVPIYAWQVAENILPDIDCKGYRIDD SYTFCFSLHKYDLIAFQKDEKSKVEFAYYINCDSSNGRFYLAWHDKGSKEQQFRISTQNLVLIQ KYQVNELGKEIRPCRLKKRPPVRGGGGSGGGGSGGGGSPAAKKKKLDGGGSKRPAATKK AGQAKKKK*
9. Therapeutic Applications
Although compact Cas9 orthologs have been previously validated for genome editing, including via single-AAV delivery, their longer PAMs have restricted therapeutic development due to target site frequencies that are lower than that of the more widely adopted SpyCas9. In addition, SauCas9 and its KKH variant with relaxed PAM requirements (Kleinstiver et al., 2015) are prone to off-target editing with some sgRNAs (Friedland et al., 2015; Kleinstiver et al., 2015). These limitations are exacerbated with target loci that require editing within a narrow sequence window, or that require precise segmental deletion. We have identified Nme2Cas9 as a compact and highly accurate Cas9 with a less restrictive dinucleotide PAM for genome editing by AAV delivery in vivo. The development of Nme2Cas9 greatly expands the genomic scope of in vivo editing, especially via viral vector delivery. The Nme2Cas9 all-in-one AAV delivery platform established in this study can in principle be used to target as wide a range of sites as SpyCas9 (due to the identical densities of optimal N4CC and NGG PAMs), but without the need to deliver two separate vectors to the same target cells. The availability of a catalytically dead version of Nme2Cas9 (dNme2Cas9) also promises to expand the scope of applications such as CRISPRi, CRISPRa, base editing, and related approaches (Dominguez et al., 2016; Komor et al., 2017). Moreover, Nme2Cas9’s hyper-accuracy enables precise editing of target genes, potentially ameliorating safety issues resulting from off-target activities. Perhaps
counterintuitively, the higher target site density of Nme2Cas9 (compared to that of Nme1Cas9) does not lead to a relative increase in off-target editing for the former. Similar results have been reported recently with SpyCas9 variants evolved to have shorter PAMs (Hu et al., 2018). Type II-C Cas9 orthologs are generally slower nucleases in vitro than SpyCas9 (Ma et al., 2015; Mir et al., 2018); interestingly, enzymological principles indicate that a reduced apparent kcat (within limits) can improve on- vs. off-target specificity for RNA-guided nucleases (Bisaria et al., 2017).
The discovery of Nme2Cas9 and Nme3Cas9 hinged on unexplored Cas9s that are highly related (outside of the PID) to an ortholog that was previously validated for human genome editing (Esvelt et al., 2013; Hou et al., 2013; Lee et al., 2016; Amrani et al., 2018). The relatedness of Nme2Cas9 and Nme3Cas9 to Nme1Cas9 brought an added benefit, namely that they use the exact same sgRNA scaffold, circumventing the need to identify and validate functional tracrRNA sequences for each. In the context of natural CRISPR immunity, the accelerated evolution of novel PAM specificities could reflect selective pressure to restore targeting of phages and MGEs that have escaped interference through PAM mutations (Deveau et al., 2008; Paez-Espino et al., 2015). Our observation that AcrIIC5Smu inhibits Nme1Cas9 but not Nme2Cas9 suggests a second, non-mutually-exclusive basis for accelerated PID variation, namely evasion of anti-CRISPR inhibition. We also speculate that accelerated variability may not be restricted to PIDs, perhaps resulting from selective pressures to evade anti-CRISPRs that bind other Cas9 domains. Cas9 inhibitors such as AcrIIC1 that bind more conserved regions of Cas9 likely present fewer routes toward mutational escape and therefore exhibit a broader inhibitory spectrum (Harrington et al., 2017a). Whatever the sources of selective pressure driving Acr and Cas9 co-evolution, the availability of validated inhibitors of Nme2Cas9 (e.g. AcrIIC1-4) provides opportunities for additional levels of control over its activities. The approach used in this study (i.e. searching for rapidly-evolving domains within Cas9) can be implemented elsewhere, especially with bacterial species that are well-sampled at the level of genome sequence. This approach could also be applied to other CRISPR-Cas effector proteins such as Cas12 and Cas13 that have also been developed for genome or transcriptome engineering and other applications. This strategy could be especially compelling with Cas proteins that are closely related to orthologs with proven efficacy in heterologous contexts (e.g. in eukaryotic cells), as was the case for Nme1Cas9. The application of this approach to meningococcal Cas9 orthologs yielded a new genome editing platform, Nme2Cas9, with a unique combination of characteristics (compact size, dinucleotide PAM, hyper-accuracy, single- AAV deliverability, and Acr susceptibility) that promise to accelerate the development of genome editing tools for both general and therapeutic applications. Table 3. The following presents exemplary sequences for plasmids and oligos as disclosed herein.
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0001
Figure imgf000094_0001
RNP Delivery for mammalian genome editing
For RNP experiments, the Neon electroporation system was used exactly as described (Amrani et al., 2018). Briefly, 40 picomoles of 3xNLS-Nme2Cas9 along with 50 picomoles of T7-transcribed sgRNA was assembled in buffer R and electroporated using 10 mL Neon tips. After electroporation, cells were plated in pre-warmed 24-well plates containing the appropriate culture media without antibiotics. Electroporation parameters (voltage, width, number of pulses) were 1150 V, 20 ms, 2 pulses for HEK293T cells; 1000 V, 50 ms, 1 pulse for K562 cells.
In vivo AAV8.Nme2Cas9+sgRNA delivery and liver tissue processing For the AAV8 vector injections, 8-week-old female C57BL/6NJ mice were injected with 4 x1011 genome copies per mouse via tail vein, with the sgRNA targeting a validated site in either Pcsk9 or Rosa26. Mice were sacrificed 28 days after vector administration and liver tissues were collected for analysis. Liver tissues were fixed in 4% formalin overnight, embedded in paraffin, sectioned and stained with hematoxylin and eosin (H&E). Blood was drawn from the facial vein at 0, 14 and 28 days post injection, and serum was isolated using a serum separator (BD, Cat. No.365967) and stored at -80°C until assay. Serum cholesterol level was measured using the Infinity™ colorimetric endpoint assay (Thermo-Scientific) following the
manufacturer’s protocol and as previously described (Ibraheim et al., 2018). For the anti-PCSK9 Western blot, 40 mg of protein from tissue or 2 ng of Recombinant Mouse PCSK9 Protein (R&D Systems, 9258-SE-020) were loaded onto a MiniPROTEAN® TGX™ Precast Gel (Bio-Rad). The separated bands were transferred onto a PVDF membrane and blocked with 5% Blocking- Grade Blocker solution (Bio-Rad) for 2 hours at room temperature. Next, the membrane was incubated with rabbit anti-GAPDH (Abcam ab9485, 1:2,000) or goat anti-PCSK9 (R&D
Systems AF3985, 1:400) antibodies overnight. Membranes were washed in TBST and incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit (Bio-Rad 1706515, 1:4,000), and donkey anti-goat (R&D Systems HAF109, 1:2,000) secondary antibodies for 2 hours at room temperature. The membranes were washed again in TBST and visualized using Clarity™ western ECL substrate (Bio-Rad) using an M35A XOMAT Processor (Kodak).
Ex vivo AAV6.Nme2Cas9 delivery in mouse zygotes
Zygotes were incubated in 15 ml drops of KSOM (Potassium-Supplemented Simplex Optimized Medium, Millipore, Cat. No. MR-106-D) containing 3x109 or 3x108 GCs of
AAV6.Nme2Cas9.sgTyr vector for 5-6 h (4 zygotes in each drop). After incubation, zygotes were rinsed in M2 and transferred to fresh KSOM for overnight culture. The next day, the embryos that advanced to 2-cell stage were transferred into the oviduct of pseudopregnant recipients and allowed to develop to term. Experimental
Example I
Discovery of Cas9 orthologs with differentially diverged PIDs Nme1Cas9 peptide sequence was used as a query in BLAST searches to find all Cas9 orthologs in Neisseria meningitidis species. Orthologs with >80% identity to Nme1Cas9 were selected for the remainder of this study. The PIDs were then aligned with that of Nme1Cas9 (residues 820-1082) using ClustalW2 and those with clusters of mutations in the PID were selected for further analysis. An unrooted phylogenetic tree of NmeCas9 orthologs was constructed using FigTree (http://tree.bio.ed.ac.uk/software/figtree/).
Example II
Cloning, expression and purification of Cas9 and Acr orthologs Examples of plasmids and oligonucleotides used in this study are listed in Table 3. The PIDs of Nme2Cas9 and Nme3Cas9 were ordered as gBlocks (IDT) to replace the PID of Nme1Cas9 using Gibson Assembly (NEB) in the bacterial expression plasmid pMSCG7 (Zhang et al., 2015), which encodes Nme1Cas9 with a 6xHis tag. The construct was transformed into E. coli, expressed and purified as previously described (Pawluk et al., 2016). Briefly, Rosetta (DE3) cells containing the respective Cas9 plasmids were grown at 37°C to an OD600 of 0.6 and protein expression was induced by 1mM IPTG for 16 hr at 18°C. Cells were harvested and lysed by sonication in lysis buffer [50 mM Tris-HCl (pH 7.5), 500 mM NaCl, 5 mM imidazole, 1 mM DTT] supplemented with 1 mg/mL Lysozyme and protease inhibitor cocktail (Sigma). The lysate was then run through a Ni2+-NTA agarose column (Qiagen), and the bound protein was eluted with 300 mM imidazole and dialyzed into storage buffer [20 mM HEPES-NaOH (pH 7.5), 250 mM NaCl, 1 mM DTT]. For Acr proteins, 6xHis-tagged proteins were expressed in E. coli strain BL21 Rosetta (DE3). Cells were grown at 37 qC to an optical density (OD600) of 0.6 in a shaking incubator. The bacterial cultures were cooled to 18°C, and protein expression was induced by adding 1 mM IPTG for overnight expression. The next day, cells were harvested and resuspended in lysis buffer supplemented with 1 mg/mL Lysozyme and protease inhibitor cocktail (Sigma) and protein was purified using the same protocol as for Cas9. The 6xHis tag was removed by incubation of the resin-bound protein with Tobacco Etch Virus (TEV) protease overnight at 4°C to isolate untagged Acrs.
Example III
In vitro PAM discovery assay
A dsDNA target library with randomized PAM sequences was generated by overlapping PCR, with the forward primer containing the 10-nt randomized PAM region. The library was gel-purified and subjected to in vitro cleavage reaction by purified Cas9 along with T7- transcribed sgRNAs.300 nM Cas9:sgRNA complex was used to cleave 300 nM of the target fragment in 1X NEBuffer 3.1 (NEB) at 37˚C for 1 hr. The reaction was then treated with proteinase K at 50˚C for 10 minutes and run on a 4% agarose/1xTAE gel. The cleavage product was excised, eluted, and cloned using a previously described protocol (Zhang et al., 2012), with modifications. Briefly, DNA ends were repaired, non-templated 2’-deoxyadenosine tails were added, and Y-shaped adapters were ligated. After PCR, the product was quantitated with KAPA Library Quantification Kit and sequenced using a NextSeq 500 (Illumina) to obtain 75 nt paired- end reads. Sequences were analyzed with custom scripts and R.
Example IV
Transfections and mammalian genome editing
Human codon-optimized Nme2Cas9 was cloned by Gibson Assembly into the pCDest2 plasmid backbone previously used for Nme1Cas9 and SpyCas9 expression (Pawluk et al., 2016; Amrani et al., 2018). Transfection of HEK293T and HEK293T-TLR2.0 cells was performed as previously described (Amrani et al., 2018). For Hepa1-6 transfections, Lipofectamine LTX was used to transfect 500ng of all-in-one AAV.sgRNA.Nme2Cas9 plasmid in 24-well plates (~105 cells/well), using cells that had been cultured 24 hours before transfection. For K562 cells stably expressing Nme2Cas9 delivered via lentivector (see below), 50,000– 150,000 cells were electroporated with 500 ng sgRNA plasmid using 10 mL Neon tips. To measure indels in all cells 72 hr after transfections, cells were harvested and genomic DNA was extracted using the DNaesy Blood and Tissue kit (Qiagen). The targeted locus was amplified by PCR, Sanger- sequenced (Genewiz), and analyzed by TIDE (Brinkman et al., 2014) using the Desktop Genetics web-based interface (http://tide.deskgen.com).
Example V
Lentiviral transduction of K562 cells to stably express Nme2Cas9 K562 cells stably expressing Nme2Cas9 were generated as previously described for Nme1Cas9 (Amrani et al., 2018). For lentivirus production, the lentiviral vector was co- transfected into HEK293T cells along with the packaging plasmids (Addgene 12260 & 12259) in 6-well plates using TransIT-LT1 transfection reagent (Mirus Bio). After 24 hours, culture media was aspirated from the transfected cells and replaced with 1 mL of fresh DMEM. The next day, the supernatant containing the virus was collected and filtered through a 0.45 mm filter.10 uL of the undiluted supernatant along with 2.5 ug of Polybrene was used to transduce ~106 K562 cells in 6-well plates. The transduced cells were selected using media supplemented with 2.5 mg/mL puromycin.
Example VI
RNP Delivery for mammalian genome editing
For RNP experiments, the Neon electroporation system was used exactly as described (Amrani et al., 2018). Briefly, 40 picomoles of 3xNLS-Nme2Cas9 along with 50 picomoles of T7-transcribed sgRNA was assembled in buffer R and electroporated using 10 mL Neon tips. After electroporation, cells were plated in pre-warmed 24-well plates containing the appropriate culture media without antibiotics. Electroporation parameters (voltage, width, number of pulses) were 1150 V, 20 ms, 2 pulses for HEK293T cells; 1000 V, 50 ms, 1 pulse for K562 cells.
Example VII
GUIDE-seq
GUIDE-seq experiments were performed as described previously (Tsai et al., 2014), with minor modifications (Bolukbasi et al., 2015a). Briefly, HEK293T cells were transfected with 200 ng of Cas9 plasmid, 200 ng of sgRNA plasmid, and 7.5 pmol of annealed GUIDE-seq oligonucleotides using Polyfect (Qiagen). Alternatively, Hepa1-6 cells were transfected as described above. Genomic DNA was extracted with a DNeasy Blood and Tissue kit (Qiagen) 72 h after transfection according to the manufacturer’s protocol. Library preparation and sequencing were performed exactly as described previously (Bolukbasi et al., 2015a). For analysis, all sequences with up to ten mismatches with the target site, as well as a C in the fifth PAM position (N4CN), were considered potential off-target sites. Data were analyzed using the Bioconductor package GUIDEseq version 1.1.17 (Zhu et al., 2017).
Example VIII
Targeted deep sequencing and analysis
We used targeted deep sequencing to confirm the results of GUIDE-seq and to measure indel rates with maximal accuracy. We used two-step PCR amplification to produce DNA fragments for each on- and off-target site. For SpyCas9 editing at DS2 and DS6, we selected the top off-target sites based on GUIDE-seq read counts. For SpyCas9 editing at DS4, fewer candidate off-target sites were identified by GUIDE-seq, and only those with NGG (DS4|OT1, DS4|OT3, DS4|OT6) or NGC (DS4|OT2) PAMs were examined by sequencing. In the first step, we used locus-specific primers bearing universal overhangs with ends complementary to the adapters. In the first step, 2x PCR master mix (NEB) was used to generate fragments bearing the overhangs. In the second step, the purified PCR products were amplified with a universal forward primer and indexed reverse primers. Full-size products (~250 bp) were gel-purified and sequenced on an Illumina MiSeq in paired-end mode. MiSeq data analysis was performed as previously described (Pinello et al., 2016; Ibraheim et al., 2018).
Example IX
Off-target analysis using CRISPRseek
Global off-target predictions for TS25 and TS47 were performed using the Bioconductor package CRISPRseek. Minor changes were made to accommodate characteristics of Nme2Cas9 not shared with SpyCas9. Specifically, we used the following changes to: gRNA.size = 24, PAM = "NNNNCC", PAM.size = 6, RNA.PAM.pattern = "NNNNCN", and candidate off-target sites with fewer than 6 mismatches were collected. The top potential off-target sites based on the numbers and positions of mismatches were selected. Genomic DNA from cells targeted by each respective sgRNA was used to amplify each candidate off-target locus and then analyzed by TIDE.
Example X
Mouse strains and embryo collection All animal experiments were conducted under the guidance of the Institutional Animal Care and Use Committee (IACUC) of the University of Massachusetts Medical School. C57BL/6NJ (Stock No.005304). Mice were obtained from The Jackson Laboratory. All animals were maintained in a 12 h light cycle. The middle of the light cycle of the day when a mating plug was observed was considered embryonic day 0.5 (E0.5) of gestation. Zygotes were collected at E0.5 by tearing the ampulla with forceps and incubation in M2 medium containing hyaluronidase to remove cumulus cells.
Example XI
In vivo AAV8.Nme2Cas9+sgRNA delivery and liver tissue processing For the AAV8 vector injections, 8-week-old female C57BL/6NJ mice were injected with 4 x1011 genome copies per mouse via tail vein, with the sgRNA targeting a validated site in either Pcsk9 or Rosa26. Mice were sacrificed 28 days after vector administration and liver tissues were collected for analysis. Liver tissues were fixed in 4% formalin overnight, embedded in paraffin, sectioned and stained with hematoxylin and eosin (H&E). Blood was drawn from the facial vein at 0, 14 and 28 days post injection, and serum was isolated using a serum separator (BD, Cat. No.365967) and stored at -80°C until assay. Serum cholesterol level was measured using the Infinity™ colorimetric endpoint assay (Thermo-Scientific) following the
manufacturer’s protocol and as previously described (Ibraheim et al., 2018). For the anti-PCSK9 Western blot, 40 mg of protein from tissue or 2 ng of Recombinant Mouse PCSK9 Protein (R&D Systems, 9258-SE-020) were loaded onto a MiniPROTEAN® TGX™ Precast Gel (Bio-Rad). The separated bands were transferred onto a PVDF membrane and blocked with 5% Blocking- Grade Blocker solution (Bio-Rad) for 2 hours at room temperature. Next, the membrane was incubated with rabbit anti-GAPDH (Abcam ab9485, 1:2,000) or goat anti-PCSK9 (R&D
Systems AF3985, 1:400) antibodies overnight. Membranes were washed in TBST and incubated with horseradish peroxidase (HRP)-conjugated goat anti-rabbit (Bio-Rad 1706515, 1:4,000), and donkey anti-goat (R&D Systems HAF109, 1:2,000) secondary antibodies for 2 hours at room temperature. The membranes were washed again in TBST and visualized using Clarity™ western ECL substrate (Bio-Rad) using an M35A XOMAT Processor (Kodak).
Example XII
Ex vivo AAV6.Nme2Cas9 delivery in mouse zygotes
Zygotes were incubated in 15 ml drops of KSOM (Potassium-Supplemented Simplex Optimized Medium, Millipore, Cat. No. MR-106-D) containing 3x109 or 3x108 GCs of AAV6.Nme2Cas9.sgTyr vector for 5-6 h (4 zygotes in each drop). After incubation, zygotes were rinsed in M2 and transferred to fresh KSOM for overnight culture. The next day, the embryos that advanced to 2-cell stage were transferred into the oviduct of pseudopregnant recipients and allowed to develop to term.
REFERENCES, each of which are herein incorporated by reference in their entirety: Amrani, N., Gao, X.D., Liu, P., Edraki, A., Mir, A., Ibraheim, R., Gupta, A., Sasaki, K.E., Wu, T., Donohoue, P.D., et al. (2018). NmeCas9 is an intrinsically high-fidelity genome editing platform. BioRxiv, https://doi.org/10.1101/172650.
Barrangou, R., Fremaux, C., Deveau, H., Richards, M., Boyaval, P., Moineau, S., Romero, D.A., and Horvath, P. (2007). CRISPR provides acquired resistance against viruses in prokaryotes. Science 315, 1709-1712.
Bisaria, N., Jarmoskaite, I., and Herschlag, D. (2017). Lessons from Enzyme Kinetics Reveal Specificity Principles for RNA-Guided Nucleases in RNA Interference and CRISPR- Based Genome Editing. Cell Syst.4, 21-29.
Bolukbasi, M.F., Gupta, A., Oikemus, S., Derr, A.G., Garber, M., Brodsky, M.H., Zhu, L.J., and Wolfe, S.A. (2015a). DNA-binding-domain fusions enhance the targeting range and precision of Cas9. Nat. Methods 12, 1150-1156.
Bolukbasi, M.F., Gupta, A., and Wolfe, S.A. (2015b). Creating and evaluating accurate CRISPR-Cas9 scalpels for genomic surgery. Nat. Methods 13, 41-50.
Brinkman, E.K., Chen, T., Amendola, M., and van Steensel, B. (2014). Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res.42, e168.
Brouns, S.J., Jore, M.M., Lundgren, M., Westra, E.R., Slijkhuis, R.J., Snijders, A.P., Dickman, M.J., Makarova, K.S., Koonin, E.V., and van der Oost, J. (2008). Small CRISPR RNAs guide antiviral defense in prokaryotes. Science 321, 960-964.
Casini, A., Olivieri, M., Petris, G., Montagna, C., Reginato, G., Maule, G., Lorenzin, F., Prandi, D., Romanel, A., Demichelis, F., et al. (2018). A highly specific SpCas9 variant is identified by in vivo screening in yeast. Nat. Biotechnol.36, 265-271.
Certo, M.T., Ryu, B.Y., Annis, J.E., Garibov, M., Jarjour, J., Rawlings, D.J., and Scharenberg, A.M. (2011). Tracking genome engineering outcome at individual DNA breakpoints. Nat. Methods 8, 671-676. Chen, J.S., Dagdas, Y.S., Kleinstiver, B.P., Welch, M.M., Sousa, A.A., Harrington, L.B., Sternberg, S.H., Joung, J.K., Yildiz, A., and Doudna, J.A. (2017). Enhanced proofreading governs CRISPR-Cas9 targeting accuracy. Nature 550, 407-410.
Cho, S.W., Kim, S., Kim, J.M., and Kim, J.S. (2013). Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease. Nat. Biotechnol.31, 230-232.
Cho, S.W., Kim, S., Kim, Y., Kweon, J., Kim, H.S., Bae, S., and Kim, J.S. (2014).
Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases. Genome Res.24, 132-141.
Cong, L., Ran, F.A., Cox, D., Lin, S., Barretto, R., Habib, N., Hsu, P.D., Wu, X., Jiang, W., Marraffini, L.A., et al. (2013). Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819-823.
Deltcheva, E., Chylinski, K., Sharma, C.M., Gonzales, K., Chao, Y., Pirzada, Z.A., Eckert, M.R., Vogel, J., and Charpentier, E. (2011). CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III. Nature 471, 602-607.
Deveau, H., Barrangou, R., Garneau, J.E., Labonte, J., Fremaux, C., Boyaval, P., Romero, D.A., Horvath, P., and Moineau, S. (2008). Phage response to CRISPR-encoded resistance in Streptococcus thermophilus. J. Bacteriol.190, 1390-1400.
Dominguez, A.A., Lim, W.A., and Qi, L.S. (2016). Beyond editing: repurposing
CRISPR-Cas9 for precision genome regulation and interrogation. Nat. Rev. Mol. Cell Biol.17, 5-15.
Dong, Guo, M., Wang, S., Zhu, Y., Wang, S., Xiong, Z., Yang, J., Xu, Z., and Huang, Z. (2017). Structural basis of CRISPR-SpyCas9 inhibition by an anti-CRISPR protein. Nature 546, 436-439.
Esvelt, K.M., Mali, P., Braff, J.L., Moosburner, M., Yaung, S.J., and Church, G.M.
(2013). Orthogonal Cas9 proteins for RNA-guided gene regulation and editing. Nat. Methods 10, 1116-1121.
Fonfara, I., Le Rhun, A., Chylinski, K., Makarova, K.S., Lecrivain, A.L., Bzdrenga, J., Koonin, E.V., and Charpentier, E. (2014). Phylogeny of Cas9 determines functional
exchangeability of dual-RNA and Cas9 among orthologous type II CRISPR-Cas systems.
Nucleic Acids Res.42, 2577-2590. Friedland, A.E., Baral, R., Singhal, P., Loveluck, K., Shen, S., Sanchez, M., Marco, E., Gotta, G.M., Maeder, M.L., Kennedy, E.M., et al. (2015). Characterization of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications. Genome Biol.16, 257.
Friedrich, G., and Soriano, P. (1991). Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice. Genes Dev.5, 1513-1523.
Fu, Y., Sander, J.D., Reyon, D., Cascio, V.M., and Joung, J.K. (2014). Improving CRISPR-Cas nuclease specificity using truncated guide RNAs. Nat. Biotechnol.32, 279-284.
Gallagher, D.N., and Haber, J.E. (2018). Repair of a Site-Specific DNA Cleavage: Old- School Lessons for Cas9-Mediated Gene Editing. ACS Chem. Biol.13, 397-405.
Garneau, J.E., Dupuis, M.E., Villion, M., Romero, D.A., Barrangou, R., Boyaval, P., Fremaux, C., Horvath, P., Magadan, A.H., and Moineau, S. (2010). The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 468, 67-71.
Gasiunas, G., Barrangou, R., Horvath, P., and Siksnys, V. (2012). Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria. Proc. Natl. Acad. Sci. USA 109, E2579-2586.
Gaudelli, N.M., Komor, A.C., Rees, H.A., Packer, M.S., Badran, A.H., Bryson, D.I., and Liu, D.R. (2017). Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage. Nature 551, 464-471.
Ghanta, K., Dokshin, G., Mir, A., Krishnamurthy, P., Gneid, H., Edraki, A., Watts, J., Sontheimer, E., and Mello, C. (2018).5¢ Modifications Improve Potency and Efficacy of DNA Donors for Precision Genome Editing. Biorxiv 354480.
Gorski, S.A., Vogel, J., and Doudna, J.A. (2017). RNA-based recognition and targeting: sowing the seeds of specificity. Nat. Rev. Mol. Cell Biol.18, 215-228.
Harrington, L.B., Doxzen, K.W., Ma, E., Liu, J.J., Knott, G.J., Edraki, A., Garcia, B., Amrani, N., Chen, J.S., Cofsky, J.C., et al. (2017a). A Broad-Spectrum Inhibitor of CRISPR- Cas9. Cell 170, 1224-1233.
Harrington, L.B., Paez-Espino, D., Staahl, B.T., Chen, J.S., Ma, E., Kyrpides, N.C., and Doudna, J.A. (2017b). A thermostable Cas9 with increased lifetime in human plasma. Nat. Commun.8, 1424. Hou, Z., Zhang, Y., Propson, N.E., Howden, S.E., Chu, L.F., Sontheimer, E.J., and Thomson, J.A. (2013). Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis. Proc. Natl. Acad. Sci. USA 110, 15644-15649.
Hu, J.H., Miller, S.M., Geurts, M.H., Tang, W., Chen, L., Sun, N., Zeina, C.M., Gao, X., Rees, H.A., Lin, Z., et al. (2018). Evolved Cas9 variants with broad PAM compatibility and high DNA specificity. Nature 556, 57-63.
Hwang, W.Y., Fu, Y., Reyon, D., Maeder, M.L., Tsai, S.Q., Sander, J.D., Peterson, R.T., Yeh, J.R., and Joung, J.K. (2013). Efficient genome editing in zebrafish using a CRISPR-Cas system. Nat. Biotechnol.31, 227-229.
Hynes, A.P., Rousseau, G.M., Lemay, M.-L., Horvath, P., Romero, D.A., Fremaux, C., and Moineau, S. (2017). An anti-CRISPR from a virulent streptococcal phage inhibits
Streptococcus pyogenes Cas9. Nat. Microbiol.2, 1374-1380.
Ibraheim, R., Song, C.-Q., Mir, A., Amrani, N., Xue, W., and Sontheimer, E.J. (2018). All-in-One Adeno-associated Virus Delivery and Genome Editing by Neisseria meningitidis Cas9 in vivo. BioRxiv, https://doi.org/10.1101/295055.
Jiang, F., and Doudna, J.A. (2017). CRISPR–Cas9 Structures and Mechanisms. Annu. Rev. Biophys.46, 505-529.
Jiang, W., Bikard, D., Cox, D., Zhang, F., and Marraffini, L.A. (2013). RNA-guided editing of bacterial genomes using CRISPR-Cas systems. Nat. Biotechnol.31, 233-239.
Jinek, M., Chylinski, K., Fonfara, I., Hauer, M., Doudna, J.A., and Charpentier, E.
(2012). A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 337, 816-821.
Jinek, M., East, A., Cheng, A., Lin, S., Ma, E., and Doudna, J. (2013). RNA-programmed genome editing in human cells. eLife 2, e00471.
Karvelis, T., Gasiunas, G., Young, J., Bigelyte, G., Silanskas, A., Cigan, M., and Siksnys, V. (2015). Rapid characterization of CRISPR-Cas9 protospacer adjacent motif sequence elements. Genome Biol.16, 253.
Keeler, A.M., ElMallah, M.K., and Flotte, T.R. (2017). Gene Therapy 2017: Progress and Future Directions. Clin. Transl. Sci.10, 242-248. Kim, E., Koo, T., Park, S.W., Kim, D., Kim, K.-E., Kim, K., Cho, H.-Y., Song, D.W., Lee, K.J., Jung, M.H., et al. (2017). In vivo genome editing with a small Cas9 ortholog derived from Campylobacter jejuni. Nat. Commun.8, 14500.
Kim, S., Kim, D., Cho, S.W., Kim, J., and Kim, J.S. (2014). Highly efficient RNA- guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins. Genome Res.24, 1012-1019.
Kim, B., Komor, A., Levy, J., Packer, M., Zhao, K., and Liu, D. (2017). Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions. Nature Biotechnology 35.
Kleinstiver, B.P., Prew, M.S., Tsai, S.Q., Nguyen, N.T., Topkar, V.V., Zheng, Z., and Joung, J.K. (2015). Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition. Nat. Biotechnol.33, 1293-1298.
Kluesner, M., Nedveck, D., Lahr, W., Garbe, J., Abrahante, J., Webber, B., and
Moriarity, B. (2018). EditR: A Method to Quantify Base Editing from Sanger Sequencing. The CRISPR Journal 1, 239–250.
Koblan, L., Doman, J., Wilson, C., Levy, J., Tay, T., Newby, G., Maianti, J., Raguram, A., and Liu, D. (2018). Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction. Nat Biotechnol 36, 843.
Komor, A.C., Badran, A.H., and Liu, D.R. (2017). CRISPR-Based Technologies for the Manipulation of Eukaryotic Genomes. Cell 168, 20-36.
Komor, A.C., Kim, Y.B., Packer, M.S., Zuris, J.A., and Liu, D.R. (2016). Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature 533, 420-424.
Lee, C.M., Cradick, T.J., and Bao, G. (2016). The Neisseria meningitidis CRISPR-Cas9 system enables specific genome editing in mammalian cells. Mol. Ther.24, 645-654.
Lee, J., Mir, A., Edraki, A., Garcia, B., Amrani, N., Lou, H.E., Gainetdinov, I., Pawluk, A., Ibraheim, R., Gao, X.D., et al. (2018). Potent Cas9 inhibition in bacterial and human cells by new anti-CRISPR protein families. BioRxiv,
https://www.biorxiv.org/content/early/2018/2006/2020/350504.
Ma, E., Harrington, L.B., O'Connell, M.R., Zhou, K., and Doudna, J.A. (2015). Single- Stranded DNA Cleavage by Divergent CRISPR-Cas9 Enzymes. Mol. Cell 60, 398-407. Mali, P., Aach, J., Stranges, P.B., Esvelt, K.M., Moosburner, M., Kosuri, S., Yang, L., and Church, G.M. (2013a). CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering. Nat. Biotechnol.31, 833-838.
Mali, P., Yang, L., Esvelt, K.M., Aach, J., Guell, M., DiCarlo, J.E., Norville, J.E., and Church, G.M. (2013b). RNA-guided human genome engineering via Cas9. Science 339, 823- 826.
Marraffini, L.A., and Sontheimer, E.J. (2008). CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA. Science 322, 1843-1845.
Mir, A., Edraki, A., Lee, J., and Sontheimer, E.J. (2018). Type II-C CRISPR-Cas9 biology, mechanism and application. ACS Chem. Biol.13, 357-365.
Mojica, F.J., Diez-Villasenor, C., Garcia-Martinez, J., and Almendros, C. (2009). Short motif sequences determine the targets of the prokaryotic CRISPR defence system. Microbiology 155, 733-740.
Paez-Espino, D., Sharon, I., Morovic, W., Stahl, B., Thomas, B.C., Barrangou, R., and Banfield, J.F. (2015). CRISPR immunity drives rapid phage genome evolution in Streptococcus thermophilus. mBio 6.
Pawluk, A., Amrani, N., Zhang, Y., Garcia, B., Hidalgo-Reyes, Y., Lee, J., Edraki, A., Shah, M., Sontheimer, E.J., Maxwell, K.L., et al. (2016). Naturally occurring off-switches for CRISPR-Cas9. Cell 167, 1829-1838 e1829.
Pawluk, A., Bondy-Denomy, J., Cheung, V.H., Maxwell, K.L., and Davidson, A.R.
(2014). A new group of phage anti-CRISPR genes inhibits the type I-E CRISPR-Cas system of Pseudomonas aeruginosa. mBio 5, e00896.
Pinello, L., Canver, M.C., Hoban, M.D., Orkin, S.H., Kohn, D.B., Bauer, D.E., and Yuan, G.C. (2016). Analyzing CRISPR genome-editing experiments with CRISPResso. Nat.
Biotechnol.34, 695-697.
Racanelli, V., and Rehermann, B. (2006). The liver as an immunological organ.
Hepatology 43, S54-62.
Ran, F.A., Cong, L., Yan, W.X., Scott, D.A., Gootenberg, J.S., Kriz, A.J., Zetsche, B., Shalem, O., Wu, X., Makarova, K.S., et al. (2015). In vivo genome editing using Staphylococcus aureus Cas9. Nature 520, 186-191. Ran, F.A., Hsu, P.D., Lin, C.Y., Gootenberg, J.S., Konermann, S., Trevino, A.E., Scott, D.A., Inoue, A., Matoba, S., Zhang, Y., et al. (2013). Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity. Cell 154, 1380-1389.
Rashid, S., Curtis, D.E., Garuti, R., Anderson, N.N., Bashmakov, Y., Ho, Y.K., Hammer, R.E., Moon, Y.A., and Horton, J.D. (2005). Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9. Proc. Natl. Acad. Sci. USA 102, 5374-5379.
Rauch, B.J., Silvis, M.R., Hultquist, J.F., Waters, C.S., McGregor, M.J., Krogan, N.J., and Bondy-Denomy, J. (2017). Inhibition of CRISPR-Cas9 with Bacteriophage Proteins. Cell 168, 150-158 e110.
Sapranauskas, R., Gasiunas, G., Fremaux, C., Barrangou, R., Horvath, P., and Siksnys, V. (2011). The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli. Nucleic Acids Res.39, 9275-9282.
Schumann, K., Lin, S., Boyer, E., Simeonov, D.R., Subramaniam, M., Gate, R.E., Haliburton, G.E., Ye, C.J., Bluestone, J.A., Doudna, J.A., et al. (2015). Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc. Natl. Acad. Sci. USA 112, 10437- 10442.
Shin, J., Jiang, F., Liu, J.J., Bray, N.L., Rauch, B.J., Baik, S.H., Nogales, E., Bondy- Denomy, J., Corn, J.E., and Doudna, J.A. (2017). Disabling Cas9 by an anti-CRISPR DNA mimic. Sci. Adv.3, e1701620.
Tsai, S.Q., and Joung, J.K. (2016). Defining and improving the genome-wide specificities of CRISPR-Cas9 nucleases. Nat. Rev. Genet.17, 300-312.
Tsai, S.Q., Zheng, Z., Nguyen, N.T., Liebers, M., Topkar, V.V., Thapar, V., Wyvekens, N., Khayter, C., Iafrate, A.J., Le, L.P., et al. (2014). GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases. Nat. Biotechnol.33, 187-197.
Tycko, J., Myer, V.E., and Hsu, P.D. (2016). Methods for optimizing CRISPR-Cas9 genome editing specificity. Mol. Cell 63, 355-370.
Yang, H., and Patel, D.J. (2017). Inhibition Mechanism of an Anti-CRISPR Suppressor AcrIIA4 Targeting SpyCas9. Mol Cell 67, 117-127 e115.
Yin, H., Song, C.Q., Suresh, S., Kwan, S.Y., Wu, Q., Walsh, S., Ding, J., Bogorad, R.L., Zhu, L.J., Wolfe, S.A., et al. (2018). Partial DNA-guided Cas9 enables genome editing with reduced off-target activity. Nat. Chem. Biol.14, 311-316. Yokoyama, T., Silversides, D.W., Waymire, K.G., Kwon, B.S., Takeuchi, T., and Overbeek, P.A. (1990). Conserved cysteine to serine mutation in tyrosinase is responsible for the classical albino mutation in laboratory mice. Nucleic Acids Res.18, 7293-7298.
Yoon, Y., Wang, D., Tai, P.W.L., Riley, J., Gao, G., and Rivera-Perez, J.A. (2018).
Streamlined ex vivo and in vivo genome editing in mouse embryos using recombinant adeno- associated viruses. Nat. Commun.9, 412.
Zhang, Y., Heidrich, N., Ampattu, B.J., Gunderson, C.W., Seifert, H.S., Schoen, C., Vogel, J., and Sontheimer, E.J. (2013). Processing-independent CRISPR RNAs limit natural transformation in Neisseria meningitidis. Mol. Cell 50, 488-503.
Zhang, Y., Rajan, R., Seifert, H.S., Mondragón, A., and Sontheimer, E.J. (2015). DNase H activity of Neisseria meningitidis Cas9. Mol. Cell 60, 242-255.
Zhang, Z., Theurkauf, W.E., Weng, Z., and Zamore, P.D. (2012). Strand-specific libraries for high throughput RNA sequencing (RNA-Seq) prepared without poly(A) selection. Silence 3, 9.
Zhu, L.J., Holmes, B.R., Aronin, N., and Brodsky, M.H. (2014). CRISPRseek: a bioconductor package to identify target-specific guide RNAs for CRISPR-Cas9 genome-editing systems. PLoS One 9, e108424.
Zhu, L.J., Lawrence, M., Gupta, A., Pagés, H., Kucukural, A., Garber, M., and Wolfe, S.A. (2017). GUIDEseq: a bioconductor package to analyze GUIDE-Seq datasets for CRISPR- Cas nucleases. BMC Genomics 18, 379.
Zuris, J.A., Thompson, D.B., Shu, Y., Guilinger, J.P., Bessen, J.L., Hu, J.H., Maeder, M.L., Joung, J.K., Chen, Z.-Y., and Liu, D.R. (2015). Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo. Nat. Biotechnol.33, 73-80.
All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described methods and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in biological control, biochemistry, molecular biology, entomology, plankton, fishery systems, and fresh water ecology, or related fields are intended to be within the scope of the following claims.

Claims

Claims
We claim: 1. A mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N4CC nucleotide sequence.
2. The protein of Claim 1, wherein said protein is Nme2Cas9.
3. The protein of Claim 1, further comprising a nuclear localization signal protein.
4. The protein of Claim 1, wherein said nucleotide deaminase is a cytidine deaminase.
5. The protein of Claim 1, wherein said nucleotide deaminase is an adenosine deaminase.
6. The protein of Claim 1, further comprising a uracil glycosylase inhibitor.
7. The protein of Claim 1, wherein said nuclear localization signal protein is selected from a nucleoplasmin and an SV40.
8. The protein of Claim 1, wherein said binding region is a protospacer accessory motif interacting domain.
9. The protein of Claim 8, wherein said protospacer accessory motif interacting domain comprises said mutation.
10. The protein of Claim 9, wherein said mutation is a D16A mutation.
11. An adeno-associated virus comprising a mutated NmeCas9 protein, said mutated
NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for an N4CC nucleotide sequence.
12. The virus of Claim 11, wherein said virus is an adeno-associated virus 8.
13. The virus of Claim 11, wherein said virus is an adeno-associated virus 6.
14. The virus of Claim 11, wherein said protein is Nme2Cas9.
15. The virus of Claim 11, wherein said protein further comprising a nuclear localization signal protein.
16. The virus of Claim 11, wherein said nucleotide deaminase is a cytidine deaminase.
17. The virus of Claim 11, wherein said nucleotide deaminase is an adenosine deaminase.
18. The virus of Claim 11, wherein said protein further comprises a uracil glycosylase
inhibitor.
19. The virus of Claim 11, wherein said nuclear localization signal protein is selected from a nucleoplasmin and SV40.
20. The virus of Claim 11, wherein said binding region is a protospacer accessory motif interacting domain.
21. The virus of Claim 20, wherein said protospacer accessory motif interacting domain comprises said mutation.
22. The virus of Claim 21, wherein said mutation is a D16A mutation.
23. A method, comprising:
a) providing;
i) a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence; ii) a mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence;
b) contacting said nucleotide sequence with said mutated NmeCas9 protein under conditions such that said binding region attaches to said N4CC nucleotide sequence; and
c) replacing said mutated single base with a wild type base with said mutated
NmeCas9 protein.
24. The method of Claim 23, wherein said protein is Nme2Cas9.
25. The method of Claim 23, wherein said protein further comprising a nuclear localization signal protein.
26. The method of Claim 23, wherein said nucleotide deaminase is a cytidine deaminase.
27. The method of Claim 23, wherein said nucleotide deaminase is an adenosine deaminase.
28. The method of Claim 23, wherein said protein further comprises a uracil glycosylase inhibitor.
29. The method of Claim 23, wherein said nuclear localization signal protein is selected from the group consisting of nucleoplasmin and SV40.
30. The method of Claim 23, wherein said binding region is a protospacer accessory motif interacting domain.
31. The method of Claim 30, wherein said protospacer accessory motif interacting domain comprises said Cas9 protein mutation.
32. The method of Claim 31, wherein said Cas9 protein mutation is a D16A mutation.
33. A method, comprising:
a) providing;
i) a patient comprising a nucleotide sequence comprising a gene with a mutated single base, wherein said gene is flanked by an N4CC nucleotide sequence, wherein said mutated gene causes a genetically-based medical condition;
ii) an adeno-associated virus comprising a mutated NmeCas9 protein, said mutated NmeCas9 protein comprising a fused nucleotide deaminase and a binding region for said N4CC nucleotide sequence;
b) treating said patient with said adeno-associated virus under conditions such that said mutated NmeCas9 protein replaces said mutated single base with a wild type single base, such that said genetically-based medical condition does not develop.
34. The method of Claim 33, wherein said gene encodes a tyrosinase protein.
35. The method of Claim 33, wherein said genetically-based medical condition is
tyrosinemia.
36. The method of Claim 33, wherein said virus is an adeno-associated virus 8.
37. The method of Claim 33, wherein said virus is an adeno-associated virus 6.
38. The method of Claim 33, wherein said protein is Nme2Cas9.
39. The method of Claim 33, wherein said protein further comprises a nuclear localization signal protein.
40. The method of Claim 33, wherein said nucleotide deaminase is a cytidine deaminase.
41. The method of Claim 33, wherein said nucleotide deaminase is an adenosine deaminase.
42. The method of Claim 33, wherein said protein further comprises a uracil glycosylase inhibitor.
43. The method of Claim 33, wherein said nuclear localization signal protein is selected from the group consisting of nucleoplasmin and SV40.
44. The method of Claim 33, wherein said binding region is a protospacer accessory motif interacting domain.
45. The method of Claim 44, wherein said protospacer accessory motif interacting domain comprises said mutation.
46. The method of Claim 45, wherein said mutation is a D16A mutation.
PCT/US2019/056341 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins WO2020081568A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
BR112021007123-7A BR112021007123A2 (en) 2018-10-15 2019-10-15 base editing of programmable DNA by the nme2cas9-deaminase fusion proteins
MX2021004301A MX2021004301A (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins.
SG11202103722TA SG11202103722TA (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins
CN201980077403.7A CN113166743A (en) 2018-10-15 2019-10-15 Programmable DNA base editing of NME2CAS 9-deaminase fusion protein
EP19806344.8A EP3867367A1 (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins
CA3116555A CA3116555A1 (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins
AU2019362874A AU2019362874A1 (en) 2018-10-15 2019-10-15 Programmable DNA base editing by Nme2Cas9-deaminase fusion proteins
US17/285,440 US20220290113A1 (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins
EA202191033A EA202191033A1 (en) 2018-10-15 2019-10-15 PROGRAMMED DNA BASE EDITING VIA NME2CAS9-DEAMINASE fusion proteins
KR1020217014669A KR20210077732A (en) 2018-10-15 2019-10-15 Programmable DNA base editing by NME2CAS9-deaminase fusion protein
JP2021545287A JP2022508716A (en) 2018-10-15 2019-10-15 Programmable DNA base editing with Nme2Cas9-deaminase fusion protein
IL282267A IL282267A (en) 2018-10-15 2021-04-12 Programmable dna base editing by nme2cas9-deaminase fusion proteins
CONC2021/0006336A CO2021006336A2 (en) 2018-10-15 2021-05-14 Programmable DNA base editing using nme2cas9-deaminase fusion proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862745666P 2018-10-15 2018-10-15
US62/745,666 2018-10-15

Publications (1)

Publication Number Publication Date
WO2020081568A1 true WO2020081568A1 (en) 2020-04-23

Family

ID=68621349

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/056341 WO2020081568A1 (en) 2018-10-15 2019-10-15 Programmable dna base editing by nme2cas9-deaminase fusion proteins

Country Status (14)

Country Link
US (1) US20220290113A1 (en)
EP (1) EP3867367A1 (en)
JP (1) JP2022508716A (en)
KR (1) KR20210077732A (en)
CN (1) CN113166743A (en)
AU (1) AU2019362874A1 (en)
BR (1) BR112021007123A2 (en)
CA (1) CA3116555A1 (en)
CO (1) CO2021006336A2 (en)
EA (1) EA202191033A1 (en)
IL (1) IL282267A (en)
MX (1) MX2021004301A (en)
SG (1) SG11202103722TA (en)
WO (1) WO2020081568A1 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022008466A1 (en) 2020-07-06 2022-01-13 Wageningen Universiteit Base editing tools
WO2022125982A1 (en) 2020-12-11 2022-06-16 Intellia Therapeutics, Inc. Compositions and methods for reducing mhc class ii in a cell
WO2022125968A1 (en) 2020-12-11 2022-06-16 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing involving deamination
WO2022140586A2 (en) 2020-12-23 2022-06-30 Intellia Therapeutics, Inc. Compositions and methods for reducing hla-a in a cell
WO2022140587A1 (en) 2020-12-23 2022-06-30 Intellia Therapeutics, Inc. Compositions and methods for genetically modifying ciita in a cell
WO2023081689A2 (en) 2021-11-03 2023-05-11 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing
WO2023081070A1 (en) * 2021-11-02 2023-05-11 University Of Massachusetts Nme2Cas9 INLAID DOMAIN FUSION PROTEINS
WO2023196802A1 (en) * 2022-04-04 2023-10-12 The Broad Institute, Inc. Cas9 variants having non-canonical pam specificities and uses thereof
WO2023245109A2 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Compositions and methods for genomic editing
WO2023245113A1 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Methods and compositions for genetically modifying a cell
WO2023245108A2 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Compositions and methods for reducing mhc class i in a cell
WO2023227669A3 (en) * 2022-05-26 2024-02-22 UCB Biopharma SRL Novel nucleic acid-editing proteins
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115206456B (en) * 2022-07-13 2023-04-25 黑龙江大学 Molecular generation method based on attribute editing stream
CN115851775B (en) * 2022-10-18 2023-08-04 哈尔滨工业大学 Cas9 protein inhibitor and application thereof
CN115716880A (en) * 2022-12-07 2023-02-28 云舟生物科技(广州)股份有限公司 Nuclear localization fluorescent protein and application thereof

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US20150166980A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Fusions of cas9 domains and nucleic acid-editing domains
WO2017070633A2 (en) * 2015-10-23 2017-04-27 President And Fellows Of Harvard College Evolved cas9 proteins for gene editing
WO2018027078A1 (en) * 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
WO2018149915A1 (en) * 2017-02-15 2018-08-23 Keygene N.V. Methods of targeted genetic alteration in plant cells
WO2018152197A1 (en) * 2017-02-15 2018-08-23 Massachusetts Institute Of Technology Dna writers, molecular recorders and uses thereof
WO2018176009A1 (en) * 2017-03-23 2018-09-27 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014150624A1 (en) * 2013-03-14 2014-09-25 Caribou Biosciences, Inc. Compositions and methods of nucleic acid-targeting nucleic acids

Patent Citations (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3850752A (en) 1970-11-10 1974-11-26 Akzona Inc Process for the demonstration and determination of low molecular compounds and of proteins capable of binding these compounds specifically
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4275149A (en) 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4366241B1 (en) 1980-08-07 1988-10-18
US20150166980A1 (en) 2013-12-12 2015-06-18 President And Fellows Of Harvard College Fusions of cas9 domains and nucleic acid-editing domains
WO2017070633A2 (en) * 2015-10-23 2017-04-27 President And Fellows Of Harvard College Evolved cas9 proteins for gene editing
US20170121693A1 (en) 2015-10-23 2017-05-04 President And Fellows Of Harvard College Nucleobase editors and uses thereof
WO2018027078A1 (en) * 2016-08-03 2018-02-08 President And Fellows Of Harard College Adenosine nucleobase editors and uses thereof
WO2018149915A1 (en) * 2017-02-15 2018-08-23 Keygene N.V. Methods of targeted genetic alteration in plant cells
WO2018152197A1 (en) * 2017-02-15 2018-08-23 Massachusetts Institute Of Technology Dna writers, molecular recorders and uses thereof
WO2018176009A1 (en) * 2017-03-23 2018-09-27 President And Fellows Of Harvard College Nucleobase editors comprising nucleic acid programmable dna binding proteins

Non-Patent Citations (89)

* Cited by examiner, † Cited by third party
Title
ALEXIS C. KOMOR ET AL: "Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage", NATURE, vol. 533, no. 7603, 20 April 2016 (2016-04-20), London, pages 420 - 424, XP055551781, ISSN: 0028-0836, DOI: 10.1038/nature17946 *
ALIREZA EDRAKI ET AL: "A Compact, High-Accuracy Cas9 with a Dinucleotide PAM for In Vivo Genome Editing", MOLECULAR CELL, vol. 73, no. 4, 20 December 2018 (2018-12-20), AMSTERDAM, NL, pages 714 - 726.e4, XP055585186, ISSN: 1097-2765, DOI: 10.1016/j.molcel.2018.12.003 *
AMRANI, N.GAO, X.D.LIU, P.EDRAKI, A.MIR, A.IBRAHEIM, R.GUPTA, A.SASAKI, K.E.WU, T.DONOHOUE, P.D. ET AL.: "NmeCas9 is an intrinsically high-fidelity genome editing platform", BIORXIV, 2018, Retrieved from the Internet <URL:https://doi.org/10.1101/172650>
AYMAN EID ET AL: "CRISPR base editors: genome editing without double-stranded breaks", BIOCHEMICAL JOURNAL, vol. 475, no. 11, 11 June 2018 (2018-06-11), pages 1955 - 1964, XP055638645, ISSN: 0264-6021, DOI: 10.1042/BCJ20170793 *
BARRANGOU, R.FREMAUX, C.DEVEAU, H.RICHARDS, M.BOYAVAL, P.MOINEAU, S.ROMERO, D.A.HORVATH, P.: "CRISPR provides acquired resistance against viruses in prokaryotes", SCIENCE, vol. 315, 2007, pages 1709 - 1712, XP002428071, DOI: 10.1126/science.1138140
BENJAMIN P. KLEINSTIVER ET AL: "Engineered CRISPR-Cas9 nucleases with altered PAM specificities", NATURE, vol. 523, no. 7561, 22 June 2015 (2015-06-22), London, pages 481 - 485, XP055293257, ISSN: 0028-0836, DOI: 10.1038/nature14592 *
BETH A. ROUSSEAU ET AL: "Programmable RNA Cleavage and Recognition by a Natural CRISPR-Cas9 System from Neisseria meningitidis", MOLECULAR CELL, vol. 69, no. 5, 15 February 2018 (2018-02-15), AMSTERDAM, NL, pages 906 - 914.e4, XP055658833, ISSN: 1097-2765, DOI: 10.1016/j.molcel.2018.01.025 *
BISARIA, N.JARMOSKAITE, 1.HERSCHLAG, D.: "Lessons from Enzyme Kinetics Reveal Specificity Principles for RN -Guided Nucleases in RNA Interference and CRISPR-Based Genome Editing", CELL SYST., vol. 4, 2017, pages 21 - 29
BOLUKBASI, M.F.GUPTA, A.OIKEMUS, S.DERR, A.G.GARBER, M.BRODSKY, M.H.ZHU, L.J.WOLFE, S.A.: "DNA-binding-domain fusions enhance the targeting range and precision of Cas9", NAT. METHODS, vol. 12, 2015, pages 1150 - 1156, XP055548728, DOI: 10.1038/nmeth.3624
BOLUKBASI, M.F.GUPTA, A.WOLFE, S.A.: "Creating and evaluating accurate CRISPR-Cas9 scalpels for genomic surgery", NAT. METHODS, vol. 13, 2015, pages 41 - 50, XP055283692, DOI: 10.1038/nmeth.3684
BRINKMAN, E.K.CHEN, T.AMENDOLA, M.VAN STEENSEL, B.: "Easy quantitative assessment of genome editing by sequence trace decomposition", NUCLEIC ACIDS RES., vol. 42, 2014, pages el68
BROUNS, S.J.JORE, M.M.LUNDGREN, M.WESTRA, E.R.SLIJKHUIS, R.J.SNIJDERS, A.P.DICKMAN, M.J.MAKAROVA, K.S.KOONIN, E.V.VAN DER OOST, J.: "Small CRISPR RNAs guide antiviral defense in prokaryotes", SCIENCE, vol. 321, 2008, pages 960 - 964, XP055401088, DOI: 10.1126/science.1159689
CASINI, A.OLIVIERI, M.PETRIS, G.MONTAGNA, C.REGINATO, G.MAULE, G.LORENZIN, F.PRANDI, D.ROMANEL, A.DEMICHELIS, F. ET AL.: "A highly specific SpCas9 variant is identified by in vivo screening in yeast", NAT. BIOTECHNOL., vol. 36, 2018, pages 265 - 271, XP055619847, DOI: 10.1038/nbt.4066
CERTO, M.T.RYU, B.Y.ANNIS, J.E.GARIBOV, M.JARJOUR, J.RAWLINGS, D.J.SCHARENBERG, A.M.: "Tracking genome engineering outcome at individual DNA breakpoints", NAT. METHODS, vol. 8, 2011, pages 671 - 676
CHEN, J.S.DAGDAS, Y.S.KLEINSTIVER, B.P.WELCH, M.M.SOUSA, A.A.HARRINGTON, L.B.STERNBERG, S.H.JOUNG, J.K.YILDIZ, A.DOUDNA, J.A.: "Enhanced proofreading governs CRISPR-Cas9 targeting accuracy", NATURE, vol. 550, 2017, pages 407 - 410, XP055535415, DOI: 10.1038/nature24268
CHO, S.W.KIM, S.KIM, J.M.KIM, J.S.: "Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease", NAT. BIOTECHNOL., vol. 31, 2013, pages 230 - 232
CHO, S.W.KIM, S.KIM, Y.KWEON, J.KIM, H.S.BAE, S.KIM, J.S.: "Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases", GENOME RES., vol. 24, 2014, pages 132 - 141, XP055227885, DOI: 10.1101/gr.162339.113
CONG, L.RAN, F.A.COX, D.LIN, S.BARRETTO, R.HABIB, N.HSU, P.D.WU, X.JIANG, W.MARRAFFMI, L.A. ET AL.: "Multiplex genome engineering using CRISPR/Cas systems", SCIENCE, vol. 339, 2013, pages 819 - 823, XP055458249, DOI: 10.1126/science.1231143
DEITCHEVA, E.CHYLINSKI, K.SHARMA, C.M.GONZALES, K.CHAO, Y.PIRZADA, Z.A.ECKERT, M.R.VOGEL, J.CHARPENTIER, E.: "CRISPR RNA maturation by trans-encoded small RNA and host factor RNase III", NATURE, vol. 471, 2011, pages 602 - 607
DEVEAU, H.BARRANGOU, R.GAMEAU, J.E.LABONTE, J.FREMAUX, C.BOYAVAL, P.ROMERO, D.A.HORVATH, P.MOINEAU, S.: "Phage response to CRJSPR-encoded resistance in Streptococcus thermophilus", J. BACTERIOL., vol. 190, 2008, pages 1390 - 1400, XP002679468, DOI: 10.1128/​JB.01412-07
DOMINGUEZ, A.A.LIM, W.A.QI, L.S.: "Beyond editing: repurposing CRISPR-Cas9 for precision genome regulation and interrogation", NAT. REV. MOL. CELL BIOL., vol. 17, 2016, pages 5 - 15, XP055345154, DOI: 10.1038/nrm.2015.2
DONG, GUO, M.WANG, S.ZHU, Y.WANG, S.XIONG, Z.YANG, J.XU, Z.HUANG, Z.: "Structural basis of CRISPR-SpyCas9 inhibition by an anti-CRISPR protein", NATURE, vol. 546, 2017, pages 436 - 439, XP055452726, DOI: 10.1038/nature22377
ESVELT, K.M.MALI, P.BRAFF, J.L.MOOSBURNER, M.YAUNG, S.J.CHURCH, G.M.: "Orthogonal Cas9 proteins for RNA-guided gene regulation and editing", NAT. METHODS, vol. 10, 2013, pages 1116 - 1121, XP055128928, DOI: 10.1038/nmeth.2681
FONFARA, LLE RHUN, A.CHYLINSKI, K.MAKAROVA, K.S.LECRIVAIN, A.L.BZDRENGA, J.KOONIN, E.V.CHARPENTIER, E.: "Phylogeny of Cas9 determines functional exchangeability of dual-RNA and Cas9 among orthologous type II CRISPR-Cas systems", NUCLEIC ACIDS RES., vol. 42, 2014, pages 2577 - 2590, XP055399937, DOI: 10.1093/nar/gkt1074
FRIEDLAND, A.E.BARAL, R.SINGHAL, P.LOVELUCK, K.SHEN, S.SANCHEZ, M.MARCO, E.GOTTA, G.M.MAEDER, M.L.KENNEDY, E.M. ET AL.: "Characterization of Staphylococcus aureus Cas9: a smaller Cas9 for all-in-one adeno-associated virus delivery and paired nickase applications", GENOME BIOL., vol. 16, 2015, pages 257, XP055347837, DOI: 10.1186/s13059-015-0817-8
FRIEDRICH, G.SORIANO, P.: "Promoter traps in embryonic stem cells: a genetic screen to identify and mutate developmental genes in mice", GENES DEV., vol. 5, 1991, pages 1513 - 1523, XP000562802
FU, Y.SANDER, J.D.REYON, D.CASCIO, V.M.JOUNG, J.K.: "Improving CRISPR-Cas nuclease specificity using truncated guide RNAs", NAT. BIOTECHNOL., vol. 32, 2014, pages 279 - 284, XP055259718, DOI: 10.1038/nbt.2808
GALLAGHER, D.N.HABER, J.E.: "Repair of a Site-Specific DNA Cleavage: Old-School Lessons for Case-Mediated Gene Editing", ACS CHEM. BIOL., vol. 13, 2018, pages 397 - 405
GARNEAU, J.E.DUPUIS, M.E.VILLION, M.ROMERO, D.A.BARRANGOU, R.BOYAVAL, P.FREMAUX, C.HORVATH, P.MAGADAN, A.H.MOINEAU, S.: "The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA", NATURE, vol. 468, 2010, pages 67 - 71, XP055181397, DOI: 10.1038/nature09523
GASIUNAS, G.BARRANGOU, R.HORVATH, P.SIKSNYS, V.: "Cas9-crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria", PROC. NATL. ACAD. SCI. USA, vol. 109, 2012, pages E2579 - 2586, XP055569955, DOI: 10.1073/pnas.1208507109
GAUDELLI, N.M.KOMOR, A.C.REES, H.A.PACKER, M.S.BADRAN, A.H.BRYSON, D.I.LIU, D.R.: "Programmable base editing of A*T to G*C in genomic DNA without DNA cleavage", NATURE, vol. 551, 2017, pages 464 - 471
GHANTA, K.DOKSHIN, G.MIR, A.KRISHNAMURTHY, P.GNEID, H.EDRAKI, A.WATTS, J.SONTHEIMER, E.MELLO, C.: "5' Modifications Improve Potency and Efficacy of DNA Donors for Precision Genome Editing", BIORXIV, 2018, pages 354480
GORSKI, S.A.VOGEL, J.DOUDNA, J.A.: "RNA-based recognition and targeting: sowing the seeds of specificity", NAT. REV. MOL. CELL BIOL., vol. 18, 2017, pages 215 - 228
HARRINGTON, L.B.DOXZEN, K.W.MA, E.LIU, J.J.KNOTT, G.J.EDRAKI, A.GARCIA, B.AMRANI, N.CHEN, J.S.COFSKY, J.C. ET AL.: "A Broad-Spectrum Inhibitor of CRISPR-Cas9", CELL, vol. 170, 2017, pages 1224 - 1233
HARRINGTON, L.B.PAEZ-ESPINO, D.STAAHL, B.T.CHEN, J.S.MA, E.KYRPIDES, N.C.DOUDNA, J.A.: "A thermostable Cas9 with increased lifetime in human plasma", NAT. COMMUN., vol. 8, 2017, pages 1424, XP055536900, DOI: 10.1038/s41467-017-01408-4
HOU, Z.ZHANG, Y.PROPSON, N.E.HOWDEN, S.E.CHU, L.F.SONTHEIMER, E.J.THOMSON, J.A.: "Efficient genome engineering in human pluripotent stem cells using Cas9 from Neisseria meningitidis", PROC. NATL. ACAD. SCI. USA, vol. 110, 2013, pages 15644 - 15649, XP055118866, DOI: 10.1073/pnas.1313587110
HU, J.H.MILLER, S.M.GEURTS, M.H.TANG, W.CHEN, 1.SUN, N.ZEINA, C.M.GAO, X.REES, H.A.LIN, Z. ET AL.: "Evolved Cas9 variants with broad PAM compatibility and high DNA specificity", NATURE, vol. 556, 2018, pages 57 - 63, XP055490065, DOI: 10.1038/nature26155
HWANG, W.Y.FU, Y.REYON, D.MAEDER, M.L.TSAI, S.Q.SANDER, J.D.PETERSON, R.T.YEH, J.R.JOUNG, J.K.: "Efficient genome editing in zebrafish using a CRISPR-Cas system", NAT. BIOTECHNOL., vol. 31, 2013, pages 227 - 229, XP055540926, DOI: 10.1038/nbt.2501
HYNES, A.P.ROUSSEAU, G.M.LEMAY, M.-L.HORVATH, P.ROMERO, D.A.FREMAUX, C.MOINEAU, S.: "An anti-CRISPR from a virulent streptococcal phage inhibits Streptococcus pyogenes Cas9", NAT. MICROBIOL., vol. 2, 2017, pages 1374 - 1380, XP055483864, DOI: 10.1038/s41564-017-0004-7
IBRAHEIM, R.SONG, C.-Q.MIR, A.AMRANI, N.XUE, W.SONTHEIMER, E.J.: "All-in-One Adeno-associated Virus Delivery and Genome Editing by Neisseria meningitidis Cas9 in vivo", BIORXIV, 2018, Retrieved from the Internet <URL:https://doi.org/10.1101/295055>
JIANG, F.DOUDNA, J.A.: "CRISPR-Cas9 Structures and Mechanisms", ANNU. REV. BIOPHYS., vol. 46, 2017, pages 505 - 529
JIANG, W.BIKARD, D.COX, D.ZHANG, F.MARRAFFINI, L.A.: "RNA-guided editing of bacterial genomes using CRISPR-Cas systems", NAT. BIOTECHNOL., vol. 31, 2013, pages 233 - 239, XP055249123, DOI: 10.1038/nbt.2508
JINEK, M.CHYLINSKI, K.FONFARA, I.HAUER, M.DOUDNA, J.A.CHARPENTIER, E.: "A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity", SCIENCE, vol. 337, no. 6096, 2012, pages 816 - 821, XP055549487, DOI: 10.1126/science.1225829
JINEK, M.EAST, A.CHENG, A.LIN, S.MA, E.DOUDNA, J.: "RNA-programmed genome editing in human cells", ELIFE, vol. 2, 2013, pages e00471, XP002699851, DOI: 10.7554/eLife.00471
KARVELIS, T.GASIUNAS, G.YOUNG, J.BIGELYTE, G.SILANSKAS, A.CIGAN, M.SIKSNYS, V.: "Rapid characterization of CRISPR-Cas9 protospacer adjacent motif sequence elements", GENOME BIOL., vol. 16, 2015, pages 253, XP055551291, DOI: 10.1186/s13059-015-0818-7
KEELER, A.M.ELMALLAH, M.K.FLOTTE, T.R.: "Gene Therapy 2017: Progress and Future Directions", CLIN. TRANSL. SCI., vol. 10, 2017, pages 242 - 248
KIM, E.KOO, T.PARK, S.W.KIM, D.KIM, K.-E.KIM, K.CHO, H.-Y.SONG, D.W.LEE, K.J.JUNG, M.H. ET AL.: "In vivo genome editing with a small Cas9 ortholog derived from Campylobacter jejuni", NAT. COMMUN., vol. 8, 2017, pages 14500
KIM, S.KIM, D.CHO, S.W.KIM, J.KIM, J.S.: "Highly efficient RNA-guided genome editing in human cells via delivery of purified Cas9 ribonucleoproteins", GENOME RES., vol. 24, 2014, pages 1012 - 1019, XP055277723, DOI: 10.1101/gr.171322.113
KIRN, B.KOMOR, A.LEVY, J.PACKER, M.ZHAO, K.LIU, D.: "Increasing the genome-targeting scope and precision of base editing with engineered Cas9-cytidine deaminase fusions", NATURE BIOTECHNOLOGY, vol. 35, 2017, pages 35
KLEINSTIVER, B.P.PREW, M.S.TSAI, S.Q.NGUYEN, N.T.TOPKAR, V.V.ZHENG, Z.JOUNG, J.K.: "Broadening the targeting range of Staphylococcus aureus CRISPR-Cas9 by modifying PAM recognition", NAT. BIOTECHNOL., vol. 33, 2015, pages 1293 - 1298, XP055309933, DOI: 10.1038/nbt.3404
KLUESNER, M.NEDVECK, D.LAHR, W.GARBE, J.ABRAHANTE, J.WEBBER, B.MORIARITY, B.: "EditR: A Method to Quantify Base Editing from Sanger Sequencing", THE CRISPR JOURNAL, vol. 1, 2018, pages 239 - 250
KOBLAN, L.DOMAN, J.WILSON, C.LEVY, J.TAY, T.NEWBY, G.MAIANTI, J.RAGURAM, A.LIU, D.: "Improving cytidine and adenine base editors by expression optimization and ancestral reconstruction", NAT BIOTECHNOL, vol. 36, 2018, pages 843
KOMOR, A.C.BADRAN, A.H.LIU, D.R.: "CRISPR-Based Technologies for the Manipulation of Eukaryotic Genome", CELL, vol. 168, 2017, pages 20 - 36, XP002781814, DOI: 10.1016/j.cell.2016.10.044
KOMOR, A.C.KIM, Y.B.PACKER, M.S.ZURIS, J.A.LIU, D.R.: "Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage", NATURE, vol. 533, 2016, pages 420 - 424, XP055551781, DOI: 10.1038/nature17946
LEE, C.M.CRADICK, T.J.BAO, G.: "The Neisseria meningitidis CRISPR-Cas9 system enables specific genome editing in mammalian cells", MOL. THER., vol. 24, 2016, pages 645 - 654, XP055449590, DOI: 10.1038/mt.2016.8
LEE, J.MIR, A.EDRAKI, A.GARCIA, B.AMRANI, N.LOU, H.E.GAINETDINOV, I.PAWLUK, A.IBRAHEIM, R.GAO, X.D. ET AL.: "Potent Cas9 inhibition in bacterial and human cells by new anti-CRISPR protein families", BIORXIV, 2018, Retrieved from the Internet <URL:https://www.biorxiv.org/content/early/2018/2006/2020/350504>
MA, E.HARRINGTON, L.B.O'CONNELL, M.R.ZHOU, K.DOUDNA, J.A.: "Single-Stranded DNA Cleavage by Divergent CRISPR-Cas9 Enzymes", MOL. CELL, vol. 60, 2015, pages 398 - 407, XP055448872, DOI: 10.1016/j.molcel.2015.10.030
MALI, P.AACH, J.STRANGES, P.B.ESVELT, K.M.MOOSBURNER, M.KOSURI, S.YANG, L.CHURCH, G.M.: "CAS9 transcriptional activators for target specificity screening and paired nickases for cooperative genome engineering", NAT. BIOTECHNOL., vol. 31, 2013, pages 833 - 838, XP055294730, DOI: 10.1038/nbt.2675
MALI, P.YANG, L.ESVELT, K.M.AACH, J.GUELL, M.DICARLO, J.E.NORVILLE, J.E.CHURCH, G.M.: "RNA-guided human genome engineering via Cas9", SCIENCE, vol. 339, 2013, pages 823 - 826, XP055469277, DOI: 10.1126/science.1232033
MARRAFFINI, L.A.SONTHEIMER, E.J.: "CRISPR interference limits horizontal gene transfer in staphylococci by targeting DNA", SCIENCE, vol. 322, 2008, pages 1843 - 1845, XP055613684, DOI: 10.1126/science.1165771
MIR, A.EDRAKI, A.LEE, J.SONTHEIMER, E.J.: "Type II-C CRISPR-Cas9 biology, mechanism and application", ACS CHEM. BIOL., vol. 13, 2018, pages 357 - 365, XP055591347, DOI: 10.1021/acschembio.7b00855
MOJICA, F.J.DIEZ-VILLASENOR, C.GARCIA-MARTINEZ, J.ALMENDROS, C.: "Short motif sequences determine the targets of the prokaryotic CRISPR defence system", MICROBIOLOGY, vol. 155, 2009, pages 733 - 740, XP055118633, DOI: 10.1099/mic.0.023960-0
NICOLE M GAUDELLI ET AL: "Programmable base editing of A.T to G.C in genomic DNA without DNA cleavage", NATURE, MACMILLAN JOURNALS LTD, LONDON, vol. 551, no. 7681, 25 October 2017 (2017-10-25), pages 464, XP002785203, ISSN: 0028-0836, Retrieved from the Internet <URL:http://www.nature.com/articles/nature24644> DOI: 10.1038/NATURE24644 *
PAEZ-ESPINO, D.SHARON, I.MOROVIC, W.STAHL, B.THOMAS, B.C.BARRANGOU, R.BANFIELD, J.F.: "CRISPR immunity drives rapid phage genome evolution in Streptococcus thermophilus", MBIO, 2015, pages 6
PAWLUK, A.AMRANI, N.ZHANG, Y.GARCIA, B.HIDALGO-REYES, Y.LEE, J.EDRAKI, A.SHAH, M.SONTHEIMER, E.J.MAXWELL, K.L. ET AL.: "Naturally occurring off-switches for CRISPR-Cas9", CELL, vol. 167, 2016, pages 1829 - 1838 el829
PAWLUK, A.BONDY-DENOMY, J.CHEUNG, V.H.MAXWELL, K.L.DAVIDSON, A.R.: "A new group of phage anti-CRISPR genes inhibits the type I-E CRISPR-Cas system of Pseudomonas aeruginosa", MBIO, vol. 5, 2014, pages e00896, XP055474866, DOI: 10.1128/mBio.00896-14
PINELLO, L.CANVER, M.C.HOBAN, M.D.ORKIN, S.H.KOHN, D.B.BAUER, D.E.YUAN, G.C.: "Analyzing CRISPR genome-editing experiments with CRISPResso", NAT. BIOTECHNOL., vol. 34, 2016, pages 695 - 697
RACANELLI, V.REHERMANN, B.: "The liver as an immunological organ", HEPATOLOGY, vol. 43, 2006, pages 54 - 62
RAN, F.A.CONG, L.YAN, W.X.SCOTT, D.A.GOOTENBERG, J.S.KRIZ, A.I.ZETSCHE, B.SHALEM, 0.WU, X.MAKAROVA, K.S. ET AL.: "In vivo genome editing using Staphylococcus aureus Cas9", NATURE, vol. 520, 2015, pages 186 - 191, XP055484527, DOI: 10.1038/nature14299
RAN, F.A.HSU, P.D.LIN, C.Y.GOOTENBERG, J.S.KONERMANN, S.TREVINO, A.E.SCOTT, D.A.INOUE, A.MATOBA, S.ZHANG, Y. ET AL.: "Double nicking by RNA-guided CRISPR Cas9 for enhanced genome editing specificity", CELL, vol. 154, 2013, pages 1380 - 1389, XP055299681, DOI: 10.1016/j.cell.2013.08.021
RASHID, S.CURTIS, D.E.GARUTI, R.ANDERSON, N.N.BASHMAKOV, Y.HO, Y.K.HAMMER, R.E.MOON, Y.A.HORTON, J.D.: "Decreased plasma cholesterol and hypersensitivity to statins in mice lacking Pcsk9", PROC. NATL. ACAD. SCI. USA, vol. 102, 2005, pages 5374 - 5379, XP002478031, DOI: 10.1073/pnas.0501652102
RAUCH, B.J.SILVIS, MR.HULTQUIST, J.F.WATERS, C.S.MCGREGOR, M.J.KROGAN, N.J.BONDY-DENOMY, J.: "Inhibition of CRISPR-Cas9 with Bacteriophage Proteins", CELL, vol. 168, 2017, pages 150 - 158 el 10
SAPRANAUSKAS, R.GASIUNAS, G.FREMAUX, C.BARRANGOU, R.HORVATH, P.SIKSNYS, V.: "The Streptococcus thermophilus CRISPR/Cas system provides immunity in Escherichia coli", NUCLEIC ACIDS RES., vol. 39, 2011, pages 9275 - 9282, XP055265024, DOI: 10.1093/nar/gkr606
SCHUMANN, K.LIN, S.BOYER, E.SIMEONOV, D.R.SUBRAMANIAM, M.GATE, R.E.HALIBURTON, G.E.YE, C.J.BLUESTONE, J.A.DOUDNA, J.A. ET AL.: "Generation of knock-in primary human T cells using Cas9 ribonucleoproteins", PROC. NATL. ACAD. SCI. USA, vol. 112, 2015, pages 10437 - 10442, XP055277999, DOI: 10.1073/pnas.1512503112
SEIDAH ET AL.: "The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): liver regeneration and neuronal differentiation", PROC. NATL. ACAD. SCI. U.S.A., vol. 100, no. 3, 2003, pages 928 - 933, XP002495458, DOI: 10.1073/pnas.0335507100
SHIN, J.JIANG, F.LIU, J.J.BRAY, N.L.RAUCH, B.J.BAIK, S.H.NOGALES, E.BONDY-DENOMY, J.CORN, J.E.DOUDNA, J.A.: "Disabling Cas9 by an anti-CRISPR DNA mimic", SCI. ADV., vol. 3, 2017, pages el701620
TSAI, S.Q.JOUNG, J.K.: "Defining and improving the genome-wide specificities of CRISPR-Cas9 nucleases", NAT. REV. GENET., vol. 17, 2016, pages 300 - 312, XP055555620, DOI: 10.1038/nrg.2016.28
TSAI, S.Q.ZHENG, Z.NGUYEN, N.T.LIEBERS, M.TOPKAR, V.V.THAPAR, V.WYVEKENS, N.KHAYTER, C.LAFRATE, A..).LE, L.P. ET AL.: "GUIDE-seq enables genome-wide profiling of off-target cleavage by CRISPR-Cas nucleases", NAT. BIOTECHNOL., vol. 33, 2014, pages 187 - 197, XP055246459, DOI: 10.1038/nbt.3117
TYCKO, J.MYER, V.E.HSU, P.D.: "Methods for optimizing CRISPR-Cas9 genome editing specificity", MOL. CELL, vol. 63, 2016, pages 355 - 370, XP029675847, DOI: 10.1016/j.molcel.2016.07.004
YANG, H.PATEL, D.J.: "Inhibition Mechanism of an Anti-CRISPR Suppressor AcrIIA4 Targeting SpyCas9", MOL CELL, vol. 67, 2017, pages 117 - 127 el 15
YIN, H.SONG, C.Q.SURESH, S.KWAN, S.Y.WU, Q.WALSH, S.DING, J.BOGORAD, R.L.ZHU, L.J.WOLFE, S.A. ET AL.: "Partial DNA-guided Cas9 enables genome editing with reduced off-target activity", NAT. CHEM. BIOL., vol. 14, 2018, pages 311 - 316, XP055515605, DOI: 10.1038/nchembio.2559
YOKOYAMA, T.SILVERSIDES, D.W.WAYMIRE, K.G.KWON, B.S.TAKEUCHI, T.OVERBEEK, P.A.: "Conserved cysteine to serine mutation in tyrosinase is responsible for the classical albino mutation in laboratory mice", NUCLEIC ACIDS RES., vol. 18, 1990, pages 7293 - 7298
YOON, Y.WANG, D.TAI, P.W.L.RILEY, J.GAO, G.RIVERA-PEREZ, J.A.: "Streamlined ex vivo and in vivo genome editing in mouse embryos using recombinant adeno-associated viruses", NAT. COMMUN., vol. 9, 2018, pages 412
ZHANG, Y.HEIDRICH, N.AMPATTU, B.J.GUNDERSON, C.W.SEIFERT, H.S.SCHOEN, C.VOGEL, J.SONTHEIMER, E.J.: "Processing-independent CRISPR RNAs limit natural transformation in Neisseria meningitidis", MOL. CELL, vol. 50, 2013, pages 488 - 503, XP028553287, DOI: 10.1016/j.molcel.2013.05.001
ZHANG, Y.RAJAN, R.SEIFERT, H.S.MONDRAGON, A.SONTHEIMER, E.J.: "DNase H activity of Neisseria meningitidis Cas9", MOL. CELL, vol. 60, 2015, pages 242 - 255, XP055451491, DOI: 10.1016/j.molcel.2015.09.020
ZHANG, Z.THEURKAUF, W.E.WENG, Z.ZAMORE, P.D.: "Strand-specific libraries for high throughput RNA sequencing (RNA-Seq) prepared without poly(A) selection", SILENCE, vol. 3, 2012, pages 9, XP002753524, DOI: 10.1186/1758-907X-3-9
ZHU, L J.LAWRENCE, M.GUPTA, A.PAGES, H.KUCUKURAL, A.GARBER, M.WOLFE, S.A.: "GUIDEseq: a bioconductor package to analyze GUIDE-Seq datasets for CRISPR-Cas nucleases", BMC GENOMICS, vol. 18, 2017, pages 379
ZHU, L.J.HOLMES, B.R.ARONIN, N.BRODSKY, M.H.: "RISPRseek: a bioconductor package to identify target-specific guide RNAs for CRISPR-Cas9 genome-editing systems", PLOS ONE, vol. 9, 2014, pages el08424
ZURIS, J.A.THOMPSON, D.B.SHU, Y.GUILINGER, J.P.BESSEN, J.L.HU, J.H.MAEDER, M.L.JOUNG, J.K.CHEN, Z.-Y.LIU, D.R.: "Cationic lipid-mediated delivery of proteins enables efficient protein-based genome editing in vitro and in vivo", NAT. BIOTECHNOL., vol. 33, 2015, pages 73 - 80, XP055562063, DOI: 10.1038/nbt.3081

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11912985B2 (en) 2020-05-08 2024-02-27 The Broad Institute, Inc. Methods and compositions for simultaneous editing of both strands of a target double-stranded nucleotide sequence
WO2022008466A1 (en) 2020-07-06 2022-01-13 Wageningen Universiteit Base editing tools
WO2022125982A1 (en) 2020-12-11 2022-06-16 Intellia Therapeutics, Inc. Compositions and methods for reducing mhc class ii in a cell
WO2022125968A1 (en) 2020-12-11 2022-06-16 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing involving deamination
WO2022140586A2 (en) 2020-12-23 2022-06-30 Intellia Therapeutics, Inc. Compositions and methods for reducing hla-a in a cell
WO2022140587A1 (en) 2020-12-23 2022-06-30 Intellia Therapeutics, Inc. Compositions and methods for genetically modifying ciita in a cell
WO2023081070A1 (en) * 2021-11-02 2023-05-11 University Of Massachusetts Nme2Cas9 INLAID DOMAIN FUSION PROTEINS
WO2023081689A3 (en) * 2021-11-03 2023-06-08 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing
WO2023081689A2 (en) 2021-11-03 2023-05-11 Intellia Therapeutics, Inc. Polynucleotides, compositions, and methods for genome editing
WO2023196802A1 (en) * 2022-04-04 2023-10-12 The Broad Institute, Inc. Cas9 variants having non-canonical pam specificities and uses thereof
WO2023227669A3 (en) * 2022-05-26 2024-02-22 UCB Biopharma SRL Novel nucleic acid-editing proteins
WO2023245109A2 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Compositions and methods for genomic editing
WO2023245113A1 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Methods and compositions for genetically modifying a cell
WO2023245108A2 (en) 2022-06-16 2023-12-21 Intellia Therapeutics, Inc. Compositions and methods for reducing mhc class i in a cell

Also Published As

Publication number Publication date
CA3116555A1 (en) 2020-04-23
BR112021007123A2 (en) 2021-08-10
US20220290113A1 (en) 2022-09-15
EP3867367A1 (en) 2021-08-25
AU2019362874A1 (en) 2021-05-27
MX2021004301A (en) 2021-08-05
KR20210077732A (en) 2021-06-25
EA202191033A1 (en) 2021-12-21
JP2022508716A (en) 2022-01-19
IL282267A (en) 2021-05-31
CN113166743A (en) 2021-07-23
SG11202103722TA (en) 2021-05-28
CO2021006336A2 (en) 2021-10-29

Similar Documents

Publication Publication Date Title
US20220290113A1 (en) Programmable dna base editing by nme2cas9-deaminase fusion proteins
Edraki et al. A compact, high-accuracy Cas9 with a dinucleotide PAM for in vivo genome editing
US11624078B2 (en) Protected guide RNAS (pgRNAS)
US20210139872A1 (en) Crispr having or associated with destabilization domains
US20240093193A1 (en) Dead guides for crispr transcription factors
US20200389425A1 (en) Delivery, use and therapeutic applications of the crispr-cas systems and compositions for hbv and viral diseases and disorders
US10954514B2 (en) Escorted and functionalized guides for CRISPR-Cas systems
US20210222164A1 (en) Crispr-cas systems having destabilization domain
AU2018364993B2 (en) Targeted CRISPR delivery platforms
WO2023081070A1 (en) Nme2Cas9 INLAID DOMAIN FUSION PROTEINS

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19806344

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2021545287

Country of ref document: JP

Kind code of ref document: A

Ref document number: 3116555

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021007123

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20217014669

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019806344

Country of ref document: EP

Effective date: 20210517

ENP Entry into the national phase

Ref document number: 2019362874

Country of ref document: AU

Date of ref document: 20191015

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021007123

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20210414

WWE Wipo information: entry into national phase

Ref document number: 521421747

Country of ref document: SA